1
|
La Hood A, Moran C, Than S, Lu A, Collyer TA, Beare R, Srikanth V. Associations between physical activity and brain structure in a large community cohort. Sci Rep 2025; 15:18896. [PMID: 40442235 PMCID: PMC12122851 DOI: 10.1038/s41598-025-04010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 05/23/2025] [Indexed: 06/02/2025] Open
Abstract
Low physical activity (PA) and obesity are dementia risk factors, but it is unclear whether the association between PA and brain health is dependent upon degree of obesity. We aimed to examine the association between PA and brain structure and explore whether it varies by body mass index (BMI). We examined the associations between accelerometer-measured PA and magnetic resonance imaging-measured brain structure using cross-sectional data from a population-based cohort of people in midlife (UK Biobank). Using regression modelling, we explored whether these associations were mediated by BMI or other cardiometabolic risk factors. Complete data were available for 16,725 participants (median age 65 years, 55% women, median BMI 26). Greater PA was positively associated with total brain (β = 3.67, p < 0.0001), grey matter (β = 3.12, p < 0.0001), white matter (β = 0.43, p < 0.0001) and total hippocampal volumes (β = 0.05, p < 0.0001), and inversely associated with white matter hyperintensity volume (β = - 4.6 × 10-5, p < 0.0001). Although there was no interaction between PA and BMI in explaining brain volumes, BMI attenuated the PA-brain volume associations by between 18 and 58%. The PA-brain volume associations were further attenuated by other cardiometabolic risk factors and in the case of grey matter and hippocampal volume, was no longer statistically significant in fully adjusted models. Greater PA is associated with a healthier profile in brain volumes. BMI and cardiometabolic factors mediated the relationship between PA and grey matter and hippocampal volume and partially mediated its relationship with other brain volumes. These results support a mechanistic basis for PA in optimizing cardiometabolic risk for dementia prevention.
Collapse
Affiliation(s)
- Alexandra La Hood
- Peninsula Clinical School, School of Translational Medicine, Monash University, PO Box 52, Frankston, VIC, 3199, Australia
| | - Chris Moran
- Peninsula Clinical School, School of Translational Medicine, Monash University, PO Box 52, Frankston, VIC, 3199, Australia.
- Department of Geriatric Medicine, Peninsula Health, 24 Separation Street, Mornington, VIC, 3931, Australia.
- National Centre for Healthy Ageing, PO Box 52, Frankston, VIC, 3199, Australia.
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia.
- Department of Home, Acute and Community, Alfred Health, 260 Kooyong Rd, Caulfield, VIC, 3162, Australia.
| | - Stephanie Than
- Peninsula Clinical School, School of Translational Medicine, Monash University, PO Box 52, Frankston, VIC, 3199, Australia
- Department of Geriatric Medicine, Peninsula Health, 24 Separation Street, Mornington, VIC, 3931, Australia
- National Centre for Healthy Ageing, PO Box 52, Frankston, VIC, 3199, Australia
- Department of Geriatric Medicine, Western Health, 160 Gordon Street, Footscray, VIC, 3011, Australia
| | - Alicia Lu
- Peninsula Clinical School, School of Translational Medicine, Monash University, PO Box 52, Frankston, VIC, 3199, Australia
- Department of Geriatric Medicine, Peninsula Health, 24 Separation Street, Mornington, VIC, 3931, Australia
- National Centre for Healthy Ageing, PO Box 52, Frankston, VIC, 3199, Australia
| | - Taya A Collyer
- Peninsula Clinical School, School of Translational Medicine, Monash University, PO Box 52, Frankston, VIC, 3199, Australia
- National Centre for Healthy Ageing, PO Box 52, Frankston, VIC, 3199, Australia
| | - Richard Beare
- Peninsula Clinical School, School of Translational Medicine, Monash University, PO Box 52, Frankston, VIC, 3199, Australia
- National Centre for Healthy Ageing, PO Box 52, Frankston, VIC, 3199, Australia
- Developmental Imaging, Murdoch Children's Research Institute, 50 Flemington Rd, Parkville, Melbourne, VIC, 3052, Australia
| | - Velandai Srikanth
- Peninsula Clinical School, School of Translational Medicine, Monash University, PO Box 52, Frankston, VIC, 3199, Australia
- Department of Geriatric Medicine, Peninsula Health, 24 Separation Street, Mornington, VIC, 3931, Australia
- National Centre for Healthy Ageing, PO Box 52, Frankston, VIC, 3199, Australia
| |
Collapse
|
2
|
Rodrigues RS, Moreira JB, Dias P, Sebastião AM, Xapelli S. The Effects of Exercise-Associated Factors on Hippocampal Progenitor Cell Dynamics Are Mediated by Cannabinoid Type 2 Receptors. J Neurochem 2025; 169:e70091. [PMID: 40405428 DOI: 10.1111/jnc.70091] [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: 10/27/2024] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/24/2025]
Abstract
Neural stem/progenitor cells (NSPCs) operate in specialized niches of the adult mammalian brain, where their proliferative and differentiative potential is modulated by a myriad of factors. Emerging evidence sheds light on the interaction between cannabinoids and neurotrophic factors underlying a major regulatory force of NSPC dynamics. Previous data show that cannabinoid type 2 receptors (CB2Rs) tightly regulate the actions of brain-derived neurotrophic factor (BDNF), a neurotrophic factor highly upregulated during physical exercise. However, further research into the effects of exercise-associated neurotrophic factors in the regulation of NSPCs is still necessary. Therefore, we aimed at exploring the effects of exercise-associated factors in postnatal hippocampal neurogenesis and how CB2Rs regulate this process. By using dentate gyrus-derived neurospheres and treating them with a combination of exercise-associated factors, as an in vitro proxy for exercise, we found that these factors significantly promoted cell proliferation, an action partially reduced when CB2Rs were blocked. Moreover, CB2Rs were shown to be required for the actions of this exercise-mimicking cocktail in early neuronal commitment and differentiation. However, late neuronal differentiation promoted by exercise-associated factors remained unaltered in the presence of CB2R ligands. Together, these data suggest that CB2R actions are preponderant in early stages of hippocampal neurogenesis promoted by exercise. Astroglial late differentiation was also accelerated by a combination of exercise-associated factors, an effect prevented by CB2R blockage. This work provides a deeper understanding of the mechanisms underlying the actions of cannabinoids and exercise on NSPC regulation, highlighting the role of CB2R in modulating exercise-induced hippocampal neurogenesis.
Collapse
Affiliation(s)
- R S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute of Molecular Medicine, Lisbon, Portugal
| | - J B Moreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute of Molecular Medicine, Lisbon, Portugal
| | - P Dias
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute of Molecular Medicine, Lisbon, Portugal
| | - A M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute of Molecular Medicine, Lisbon, Portugal
| | - S Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute of Molecular Medicine, Lisbon, Portugal
| |
Collapse
|
3
|
Farmand S, Du Preez A, Kim C, de Lucia C, Ruepp MD, Stubbs B, Thuret S. Cognition on the move: Examining the role of physical exercise and neurogenesis in counteracting cognitive aging. Ageing Res Rev 2025; 107:102725. [PMID: 40064399 DOI: 10.1016/j.arr.2025.102725] [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: 11/29/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Structural and functional aspects of the hippocampus have been shown to be sensitive to the aging process, resulting in deficits in hippocampal-dependent cognition. Similarly, adult hippocampal neurogenesis (AHN), described as the generation of new neurons from neural stem cells in the hippocampus, has shown to be negatively affected by aging throughout life. Extensive research has highlighted the role of physical exercise (PE) in positively regulating hippocampal-dependent cognition and AHN. Here, by critically reviewing preclinical and clinical studies, we discuss the significance of PE in reversing age-associated changes of the hippocampus via modulation of AHN. We indicate that PE-induced changes operate on two main levels. On the first level, PE can potentially cause structural modifications of the hippocampus, and on the second level, it regulates the molecular and cellular pathways involved. These changes result in the vascular remodelling of the neurogenic niche, as well as the secretion of neurotrophic and antioxidant factors, which can in turn activate quiescent neural stem cells, while restoring their proliferation capacity and boosting their survival - features which are negatively impacted during aging. Understanding these mechanisms will allow us to identify new targets to tackle cognitive aging and improve quality of life.
Collapse
Affiliation(s)
- Sahand Farmand
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Curie Kim
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Chiara de Lucia
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norway
| | - Marc-David Ruepp
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; UK Dementia Research Institute at King's College London, London, United Kingdom
| | - Brendon Stubbs
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
4
|
Mostafa M, Disouky A, Lazarov O. Therapeutic modulation of neurogenesis to improve hippocampal plasticity and cognition in aging and Alzheimer's disease. Neurotherapeutics 2025; 22:e00580. [PMID: 40180804 PMCID: PMC12047516 DOI: 10.1016/j.neurot.2025.e00580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
Alzheimer's disease is characterized by progressive memory loss and cognitive decline. The hippocampal formation is the most vulnerable brain area in Alzheimer's disease. Neurons in layer II of the entorhinal cortex and the CA1 region of the hippocampus are lost at early stages of the disease. A unique feature of the hippocampus is the formation of new neurons that incorporate in the dentate gyrus of the hippocampus. New neurons form synapses with neurons in layer II of the entorhinal cortex and with the CA3 region. Immature and new neurons are characterized by high level of plasticity. They play important roles in learning and memory. Hippocampal neurogenesis is impaired early in mouse models of Alzheimer's disease and in human patients. In fact, neurogenesis is compromised in mild cognitive impairment (MCI), suggesting that rescuing neurogenesis may restore hippocampal plasticity and attenuate neuronal vulnerability and memory loss. This review will discuss the current understanding of therapies that target neurogenesis or modulate it, for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Mostafa Mostafa
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ahmed Disouky
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
5
|
Tari AR, Walker TL, Huuha AM, Sando SB, Wisloff U. Neuroprotective mechanisms of exercise and the importance of fitness for healthy brain ageing. Lancet 2025; 405:1093-1118. [PMID: 40157803 DOI: 10.1016/s0140-6736(25)00184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 04/01/2025]
Abstract
Ageing is a scientifically fascinating and complex biological occurrence characterised by morphological and functional changes due to accumulated molecular and cellular damage impairing tissue and organ function. Ageing is often accompanied by cognitive decline but is also the biggest known risk factor for Alzheimer's disease, the most common form of dementia. Emerging evidence suggests that sedentary and unhealthy lifestyles accelerate brain ageing, while regular physical activity, high cardiorespiratory fitness (CRF), or a combination of both, can mitigate cognitive impairment and reduce dementia risk. The purpose of this Review is to explore the neuroprotective mechanisms of endurance exercise and highlight the importance of CRF in promoting healthy brain ageing. Key findings show how CRF mediates the neuroprotective effects of exercise via mechanisms such as improved cerebral blood flow, reduced inflammation, and enhanced neuroplasticity. We summarise evidence supporting the integration of endurance exercise that enhances CRF into public health initiatives as a preventive measure against age-related cognitive decline. Additionally, we address important challenges such as lack of long-term studies with harmonised study designs across preclinical and clinical settings, employing carefully controlled and repeatable exercise protocols, and outline directions for future research.
Collapse
Affiliation(s)
- Atefe R Tari
- The Cardiac Exercise Research Group at the Faculty of Medicine and Health Sciences, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Trondheim, Norway
| | - Tara L Walker
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Aleksi M Huuha
- The Cardiac Exercise Research Group at the Faculty of Medicine and Health Sciences, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Trondheim, Norway
| | - Sigrid B Sando
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Trondheim, Norway
| | - Ulrik Wisloff
- The Cardiac Exercise Research Group at the Faculty of Medicine and Health Sciences, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
6
|
Li Q, Takayama N, Katsumata M, Takayama H, Kimura Y, Kumeda S, Miura T, Ichimiya T, Tan R, Shimomura H, Tateno A, Kitagawa T, Aoyagi Y, Imai M. Impacts of Forest Bathing (Shinrin-Yoku) in Female Participants with Depression/Depressive Tendencies. Diseases 2025; 13:100. [PMID: 40277811 PMCID: PMC12026234 DOI: 10.3390/diseases13040100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND It has been reported that forest bathing significantly reduced negative emotions and increased the positive feelings in both healthy males and females, as well as increasing blood serotonin in healthy males, indicating the potential for a beneficial effect on depressive status. However, an improvement effect of forest bathing on participants with depression has not been reported so far. Therefore, in order to fill this gap, this study examined the effect of forest bathing on depression in female participants with depression/depressive tendencies. METHODS Thirty-one females aged 40.1 ± 2.4 years with depression/depressive tendencies were recruited after obtaining informed consent. The study employed a randomized crossover design to compare forest bathing with city walking. They participated in day trips to a Japanese cypress forest park and to a city area of Nagano Prefecture as a control in June 2023. On both trips, they walked 2.5 km (for 90 min) in the morning and afternoon, respectively, for a total of 5.0 km per day. Blood samples were taken at 4 pm for the measurements before forest bathing on the first day and after the walking in forest and unban sites on the second and third days, at the same hospital. Concentrations of oxytocin, IGF-1, serotonin and lactic acid in blood were measured. SDS scores were calculated and the POMS test and questionnaires for subjective fatigue symptoms and sleep quality were administered before and after each trip. Temperature, humidity and illuminance were also measured in the forest and urban environments. The Nippon Medical School Central Ethics Committee approved this study. RESULTS Forest bathing significantly decreased SDS scores compared to city walk and the baseline, and the effect lasted for one week after forest bathing. Forest bathing also significantly increased the concentrations of blood serotonin in participants who were not taking antidepressants, significantly increased the levels of oxytocin and IGF-1 in blood, significantly increased the scores for positive feelings, and reduced the scores for negative emotions compared with city walking in the POMS test. In addition, forest bathing reduced subjective fatigue symptoms and improved sleep quality. CONCLUSIONS These findings provided scientific evidence to contribute to understanding forest bathing as a potential intervention for preventing depression, and future research on males should further explore these effects.
Collapse
Affiliation(s)
- Qing Li
- Department of Rehabilitation Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan; (T.K.); (Y.A.)
| | - Norimasa Takayama
- Forestry and Forest Products Research Institute, Forest Research and Management Organization, Tsukuba 300-1244, Japan;
| | - Masao Katsumata
- Nursing School, Nippon Medical School, Chiba 270-1613, Japan;
| | | | | | | | | | | | - Ruei Tan
- Tan Clinic, Tokyo 214-0001, Japan;
| | | | - Amane Tateno
- Department of Neuropsychiatry, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan;
| | - Tsunemi Kitagawa
- Department of Rehabilitation Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan; (T.K.); (Y.A.)
| | - Yoichiro Aoyagi
- Department of Rehabilitation Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan; (T.K.); (Y.A.)
| | - Michiko Imai
- INFOM (International Society of Nature and Forest Medicine), Tokyo, Japan;
| |
Collapse
|
7
|
Jeong S, K Davis C, Vemuganti R. Mechanisms of time-restricted feeding-induced neuroprotection and neuronal plasticity in ischemic stroke as a function of circadian rhythm. Exp Neurol 2025; 383:115045. [PMID: 39510297 DOI: 10.1016/j.expneurol.2024.115045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024]
Abstract
Time-restricted feeding (TRF) is known to promote longevity and brain function, and potentially prevent neurological diseases. Animal studies show that TRF enhances brain-derived neurotrophic factor (BDNF) signaling and regulates autophagy and neuroinflammation, supporting synaptic plasticity, neurogenesis and neuroprotection. Feeding/fasting paradigms influence the circadian cycle, with TRF aligning circadian cycle-related gene expression, and thus altering physiological processes. Emerging evidence highlights the role of gut microbiota in neuronal plasticity, based on the observation that TRF significantly alters gut microbiota composition. Hence, the gut-brain axis may be crucial for maintaining cognitive functions and presents a potential therapeutic target for TRF-mediated neuroprotection. In the context of ischemic stroke where neuronal damage is extensive, TRF can be a preconditioning strategy to enhance synaptic plasticity and neuronal resilience, thus improving outcomes after stroke. This review discussed the link between TRF and circadian regulation in neuronal plasticity and its implications for recovery after stroke.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin, Madison, WI, USA; William S. Middleton Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
8
|
Hiraga T, Hata T, Soya S, Shimoda R, Takahashi K, Soya M, Inoue K, Johansen JP, Okamoto M, Soya H. Light-exercise-induced dopaminergic and noradrenergic stimulation in the dorsal hippocampus: Using a rat physiological exercise model. FASEB J 2024; 38:e70215. [PMID: 39668509 PMCID: PMC11638517 DOI: 10.1096/fj.202400418rrr] [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: 02/25/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
Exercise activates the dorsal hippocampus that triggers synaptic and cellar plasticity and ultimately promotes memory formation. For decades, these benefits have been explored using demanding and stress-response-inducing exercise at moderate-to-vigorous intensities. In contrast, our translational research with animals and humans has focused on light-intensity exercise (light exercise) below the lactate threshold (LT), which almost anyone can safely perform with minimal stress. We found that even light exercise can stimulate hippocampal activity and enhance memory performance. Although the circuit mechanism of this boost remains unclear, arousal promotion even with light exercise implies the involvement of the ascending monoaminergic system that is essential to modulate hippocampal activity and impact memory. To test this hypothesis, we employed our physiological exercise model based on the LT of rats and immunohistochemically assessed the neuronal activation of the dorsal hippocampal sub-regions and brainstem monoaminergic neurons. Also, we monitored the extracellular concentration of monoamines in the dorsal hippocampus using in vivo microdialysis. We found that even light exercise increased neuronal activity in the dorsal hippocampal sub-regions and elevated the extracellular concentrations of noradrenaline and dopamine. Furthermore, we found that tyrosine hydroxylase-positive neurons in the locus coeruleus (LC) and the ventral tegmental area (VTA) were activated even by light exercise and were both positively correlated with the dorsal hippocampal activation. In conclusion, our findings demonstrate that light exercise stimulates dorsal hippocampal neurons, which are associated with LC-noradrenergic and VTA-dopaminergic activation. This shed light on the circuit mechanisms responsible for hippocampal neural activation during exercise, consequently enhancing memory function.
Collapse
Affiliation(s)
- Taichi Hiraga
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Toshiaki Hata
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Shingo Soya
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaJapan
- Department of Molecular Behavioral Physiology, Institute of MedicineUniversity of TsukubaTsukubaJapan
| | - Ryo Shimoda
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Kanako Takahashi
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Mariko Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Department of Anatomy and Neuroscience, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koshiro Inoue
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Center for Education in Liberal Arts and SciencesHealth Sciences University of HokkaidoIshikariJapan
| | - Joshua P. Johansen
- Laboratory for Neural Circuitry of MemoryRIKEN Center for Brain ScienceSaitamaJapan
| | - Masahiro Okamoto
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| |
Collapse
|
9
|
Subramanian S, Jain M, Misra R, Jain R. Peptide-based therapeutics targeting genetic disorders. Drug Discov Today 2024; 29:104209. [PMID: 39419376 DOI: 10.1016/j.drudis.2024.104209] [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: 05/24/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Genetic disorders (GDs) are challenging to treat owing to a lack of optimal treatment regimens and intricate and often difficult-to-understand underlying biological processes. Limited therapeutic approaches, which mostly provide symptomatic relief, are available. To date, a limited number of peptide-based drugs for the treatment of GDs are available, and several candidates are under clinical study. This review provides mechanistic insights into GDs and potential target areas where peptide-based drugs are beneficial. In addition, it emphasizes the usefulness of peptides as carriers for gene delivery, biomarkers for mutation detection and peptide-based vaccines for treating GDs.
Collapse
Affiliation(s)
- Shweta Subramanian
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Meenakshi Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Rajkumar Misra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S. A. S. Nagar, Punjab 160 062, India.
| |
Collapse
|
10
|
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
|
11
|
Michou V, Tsamos G, Vasdeki D, Deligiannis A, Kouidi E. Unraveling of Molecular Mechanisms of Cognitive Frailty in Chronic Kidney Disease: How Exercise Makes a Difference. J Clin Med 2024; 13:5698. [PMID: 39407758 PMCID: PMC11476541 DOI: 10.3390/jcm13195698] [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: 09/14/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
As our population ages, the medical challenges it faces become increasingly acute, with chronic kidney disease (CKD) becoming more prevalent among older adults. Frailty is alarmingly more common in CKD patients than in the general populace, putting the elderly at high risk of both physical and cognitive decline. CKD not only accelerates physical deterioration, but also heightens vascular dysfunction, calcification, arterial rigidity, systemic inflammation, oxidative stress, and cognitive impairment. Cognitive frailty, a distinct syndrome marked by cognitive deficits caused by physiological causes (excluding Alzheimer's and other dementias), is a critical concern. Although cognitive impairment has been well-studied, the molecular mechanisms driving cognitive frailty remain largely uncharted. Comprehensive interventions, including cutting-edge pharmaceuticals and lifestyle changes, are pivotal and effective, especially in the early stages of CKD. Recent research suggests that systematic exercise could counteract cognitive decline by improving brain blood flow, boosting neuroplasticity through the brain-derived neurotrophic factor (BDNF), and by triggering the release of neurotrophic factors such as insulin-like growth factor (IGF-1). This review delves into the molecular pathways of cognitive frailty in CKD, identifies key risk factors, and highlights therapeutic approaches, particularly the potent role of exercise in enhancing cognitive health.
Collapse
Affiliation(s)
- Vasiliki Michou
- Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University, 57 001 Thessaloniki, Greece; (A.D.); (E.K.)
| | - Georgios Tsamos
- Division of Endocrinology and Metabolism and Diabetes Centre, First Department of Internal Medicine, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece; (G.T.); (D.V.)
| | - Dimitra Vasdeki
- Division of Endocrinology and Metabolism and Diabetes Centre, First Department of Internal Medicine, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece; (G.T.); (D.V.)
| | - Asterios Deligiannis
- Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University, 57 001 Thessaloniki, Greece; (A.D.); (E.K.)
| | - Evangelia Kouidi
- Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University, 57 001 Thessaloniki, Greece; (A.D.); (E.K.)
| |
Collapse
|
12
|
Kim J, Suh SI, Park YJ, Kang M, Chung SJ, Lee ES, Jung HN, Eo JS, Koh SB, Oh K, Kang SH. Sarcopenia is a predictor for Alzheimer's continuum and related clinical outcomes. Sci Rep 2024; 14:21074. [PMID: 39256402 PMCID: PMC11387779 DOI: 10.1038/s41598-024-62918-y] [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: 02/29/2024] [Accepted: 05/22/2024] [Indexed: 09/12/2024] Open
Abstract
Low body mass index is closely related to a high risk of Alzheimer's disease (AD) and related biomarkers including amyloid-β (Aβ) deposition. However, the association between sarcopenia and Aβ-confirmed AD remains controversial. Therefore, we investigated the relationship between sarcopenia and the AD continuum. We explored sarcopenia's association with clinical implications of participants on the AD continuum. We prospectively enrolled 142 participants on the AD continuum (19 with preclinical AD, 96 with mild cognitive impairment due to AD, and 28 with AD dementia) and 58 Aβ-negative cognitively unimpaired participants. Sarcopenia, assessed using dual-energy X-ray absorptiometry and hand grip measurements, was considered a predictor. AD continuum, defined by Aβ deposition on positron emission tomography served as an outcome. Clinical severity in participants on the AD continuum assessed using hippocampal volume, Mini-Mental State Examination (MMSE), Seoul Verbal Learning Test (SVLT), and Clinical Dementia Rating Scale Sum of Boxes Scores (CDR-SOB) were also considered an outcome. Sarcopenia (odds ratio = 4.99, p = 0.004) was associated independently with the AD continuum after controlling for potential confounders. Moreover, sarcopenia was associated with poor downstream imaging markers (decreased hippocampal volume, β = - 0.206, p = 0.020) and clinical outcomes (low MMSE, β = - 1.364, p = 0.025; low SVLT, β = - 1.077, p = 0.025; and high CDR-SOB scores, β = 0.783, p = 0.022) in participants on the AD continuum. Sarcopenia was associated with the AD continuum and poor clinical outcome in individuals with AD continuum. Therefore, our results provide evidence for future studies to confirm whether proper management of sarcopenia can effective strategies are required for sarcopenia management to prevent the AD continuum and its clinical implications.
Collapse
Affiliation(s)
- Jeonghun Kim
- Korea Testing Laboratory, Bio and Medical Health Division, Seoul, Korea
| | - Sang-Il Suh
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Yu Jeong Park
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Korea
| | - Minwoong Kang
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Su Jin Chung
- Department of Neurology, Inje University Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Eun Seong Lee
- Department of Nuclear Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hye Na Jung
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jae Seon Eo
- Department of Nuclear Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Seong-Beom Koh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Korea
| | - Kyungmi Oh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Korea.
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Korea.
| |
Collapse
|
13
|
Blume GR, Royes LFF. Peripheral to brain and hippocampus crosstalk induced by exercise mediates cognitive and structural hippocampal adaptations. Life Sci 2024; 352:122799. [PMID: 38852798 DOI: 10.1016/j.lfs.2024.122799] [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: 02/28/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Endurance exercise leads to robust increases in memory and learning. Several exercise adaptations occur to mediate these improvements, including in both the hippocampus and in peripheral organs. Organ crosstalk has been becoming increasingly more present in exercise biology, and studies have shown that peripheral organs can communicate to the hippocampus and mediate hippocampal changes. Both learning and memory as well as other hippocampal functional-related changes such as neurogenesis, cell proliferation, dendrite morphology and synaptic plasticity are controlled by these exercise responsive peripheral proteins. These peripheral factors, also called exerkines, are produced by several organs including skeletal muscle, liver, adipose tissue, kidneys, adrenal glands and circulatory cells. Previous reviews have explored some of these exerkines including muscle-derived irisin and cathepsin B (CTSB), but a full picture of peripheral to hippocampus crosstalk with novel exerkines such as selenoprotein 1 (SEPP1) and platelet factor 4 (PF4), or old overlooked ones such as lactate and insulin-like growth factor 1 (IGF-1) is still missing. We provide 29 different studies of 14 different exerkines that crosstalk with the hippocampus. Thus, the purpose of this review is to explore peripheral exerkines that have shown to exert hippocampal function following exercise, demonstrating their particular effects and molecular mechanisms in which they could be inducing adaptations.
Collapse
Affiliation(s)
| | - Luiz Fernando Freire Royes
- Center in Natural and Exact Sciences, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil; Physical Education and Sports Center, Department of Sports Methods and Techniques, Exercise Biochemistry Laboratory (BIOEX), Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
14
|
Alghadir AH, Gabr SA, Iqbal A. Enhancing cognitive performance and mitigating dyslipidemia: the impact of moderate aerobic training on sedentary older adults. BMC Geriatr 2024; 24:678. [PMID: 39138393 PMCID: PMC11323678 DOI: 10.1186/s12877-024-05276-8] [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: 01/09/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The present study aimed to evaluate the effects of 24 weeks of moderate aerobic exercise on lipids and lipoprotein levels; Lipo (a) markers, and their association with cognitive performance in healthy older adults. METHODS A total of 150 healthy subjects (100 males and 50 females; age range: 65-95 years) were recruited for this study. Based on the LOTCA test score, subjects were classified into two groups: the control group (n = 50) and the cognitive impairment group (n = 100). Cognitive functioning, leisure-time physical activity (LTPA), lipid profile, total cholesterol, TG, HDL-c, LDL-C, and lipo(a) were assessed at baseline and post-24-week aerobic exercise interventions using LOTCA battery, pre-validated Global Physical Activity Questionnaire (GPAQ) version II, colorimetric, and immunoassay techniques, respectively. RESULTS Significant improvements in cognitive function and modulation in lipid profile and lipoprotein (a) markers were reported in all older subjects following 24 weeks of moderate exercise. LOTCA-7-sets scores significantly correlated with physical activity status and the regulation of lipids and Lipo (a) markers. Physically active persons showed higher cognitive performance along with a reduction in the levels of T-Cholest., TG, LDL-C, Lipo (a), and an increase in the levels of HDL-C and aerobic fitness VO2max compared with sedentary participants. Cognitive performance correlated positively with increased aerobic fitness, HDL-C, and negatively with T-Cholest., TG, LDL-C, and Lipo (a). However, a significant increase in the improvement of motor praxis, vasomotor organization, thinking operations, attention, and concentration were reported among older adults. CONCLUSIONS The study findings revealed that supervised moderate aerobic training for 24 weeks significantly enhances cognitive functions via mitigating older adults' lipid profiles and lipoprotein (a). Cognitive performance is positively correlated with aerobic fitness and HDL-C level and negatively with T-Cholest., TH, LDL-C, and Lipo (a).
Collapse
Affiliation(s)
- Ahmad H Alghadir
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Sami A Gabr
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Amir Iqbal
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia.
| |
Collapse
|
15
|
Li Z, Wu X, Yan Z, Cui Y, Liu Y, Cui S, Wang Y, Liu T. Unveiling the muscle-brain axis: A bidirectional mendelian randomization study investigating the causal relationship between sarcopenia-related traits and brain aging. Arch Gerontol Geriatr 2024; 123:105412. [PMID: 38513381 DOI: 10.1016/j.archger.2024.105412] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Observational studies suggest an association between sarcopenia-related traits and brain aging, but whether this association reflects a causal relationship remains unclear. This study aims to employ Mendelian randomization (MR) methods to investigate the causal impact of sarcopenia-related traits on brain aging. METHODS This study presents a comprehensive analysis of genome-wide association study (GWAS) summary data associated with sarcopenia-related traits. The data were derived from a large-scale cohort, encompassing measures such as grip strength, lean body mass, and walking pace. Measurements of brain aging were obtained from neuroimaging genetics, utilizing meta-analysis (ENIGMA) to combine magnetic resonance imaging (MRI) data from 33,992 participants. The primary methodology employed in this analysis was the inverse-variance-weighted method (IVW). Additionally, sensitivity analyses were conducted, to assess heterogeneity and pleiotropy. RESULT Appendicular lean mass(ALM) is negatively correlated with Pallidum aging; Whole body fat-free mass shows a negative correlation with Amygdala aging; Leg fat-free mass (left) and Leg fat-free mass (right) are negatively correlated with Pallidum aging; Usual walking pace is positively correlated with Nucleus Accumbens aging. Cerebellum WM aging is negatively correlated with Leg fat-free mass (left) and Leg fat-free mass (right); Hippocampus aging is negatively correlated with Hand grip strength (left) and Hand grip strength (right). Ventricles aging is positively correlated with Usual walking pace; Nucleus Accumbens aging is positively correlated with Leg fat-free mass (left) and Leg fat-free mass (right); Putamen aging is positively correlated with ALM. CONCLUSION Our study confirms that reduced muscle mass speeds up brain aging. Walking too fast raises the risk of brain aging, while maintaining or increasing appendicular lean mass, overall muscle mass, and muscle mass in both legs lowers the risk of brain aging.
Collapse
Affiliation(s)
- Zefang Li
- Department of The First Clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xueqiang Wu
- Department of Health Science, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Zhaojun Yan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine,Jinan, China.
| | - Yiping Cui
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yueling Liu
- School of mental health and psychological science, Anhui Medical University,Hefei, China
| | - Song Cui
- Department of The First Clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yining Wang
- Department of The First Clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyu Liu
- Department of The First Clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
16
|
Methi A, Islam MR, Kaurani L, Sakib MS, Krüger DM, Pena T, Burkhardt S, Liebetanz D, Fischer A. A Single-Cell Transcriptomic Analysis of the Mouse Hippocampus After Voluntary Exercise. Mol Neurobiol 2024; 61:5628-5645. [PMID: 38217668 PMCID: PMC11249425 DOI: 10.1007/s12035-023-03869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/29/2023] [Indexed: 01/15/2024]
Abstract
Exercise has been recognized as a beneficial factor for cognitive health, particularly in relation to the hippocampus, a vital brain region responsible for learning and memory. Previous research has demonstrated that exercise-mediated improvement of learning and memory in humans and rodents correlates with increased adult neurogenesis and processes related to enhanced synaptic plasticity. Nevertheless, the underlying molecular mechanisms are not fully understood. With the aim to further elucidate these mechanisms, we provide a comprehensive dataset of the mouse hippocampal transcriptome at the single-cell level after 4 weeks of voluntary wheel-running. Our analysis provides a number of interesting observations. For example, the results suggest that exercise affects adult neurogenesis by accelerating the maturation of a subpopulation of Prdm16-expressing neurons. Moreover, we uncover the existence of an intricate crosstalk among multiple vital signaling pathways such as NF-κB, Wnt/β-catenin, Notch, and retinoic acid (RA) pathways altered upon exercise in a specific cluster of excitatory neurons within the Cornu Ammonis (CA) region of the hippocampus. In conclusion, our study provides an important resource dataset and sheds further light on the molecular changes induced by exercise in the hippocampus. These findings have implications for developing targeted interventions aimed at optimizing cognitive health and preventing age-related cognitive decline.
Collapse
Affiliation(s)
- Aditi Methi
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Md Rezaul Islam
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - M Sadman Sakib
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Dennis M Krüger
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Bioinformatics Unit, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Tonatiuh Pena
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Bioinformatics Unit, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Susanne Burkhardt
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - David Liebetanz
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany.
- Department for Psychiatry and Psychotherapy, University Medical Center of Göttingen, Georg-August University, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen, Germany.
| |
Collapse
|
17
|
Dause TJ, Denninger JK, Osap R, Walters AE, Rieskamp JD, Kirby ED. Autocrine VEGF drives neural stem cell proximity to the adult hippocampus vascular niche. Life Sci Alliance 2024; 7:e202402659. [PMID: 38631901 PMCID: PMC11024344 DOI: 10.26508/lsa.202402659] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
The vasculature is a key component of adult brain neural stem cell (NSC) niches. In the adult mammalian hippocampus, NSCs reside in close contact with a dense capillary network. How this niche is maintained is unclear. We recently found that adult hippocampal NSCs express VEGF, a soluble factor with chemoattractive properties for vascular endothelia. Here, we show that global and NSC-specific VEGF loss led to dissociation of NSCs and their intermediate progenitor daughter cells from local vasculature. Surprisingly, though, we found no changes in local vascular density. Instead, we found that NSC-derived VEGF supports maintenance of gene expression programs in NSCs and their progeny related to cell migration and adhesion. In vitro assays revealed that blockade of VEGF receptor 2 impaired NSC motility and adhesion. Our findings suggest that NSCs maintain their own proximity to vasculature via self-stimulated VEGF signaling that supports their motility towards and/or adhesion to local blood vessels.
Collapse
Affiliation(s)
- Tyler J Dause
- Department of Psychology, College of Arts and Sciences, The Ohio State University, Columbus, OH, USA
| | - Jiyeon K Denninger
- Department of Psychology, College of Arts and Sciences, The Ohio State University, Columbus, OH, USA
| | - Robert Osap
- Department of Psychology, College of Arts and Sciences, The Ohio State University, Columbus, OH, USA
| | - Ashley E Walters
- Department of Psychology, College of Arts and Sciences, The Ohio State University, Columbus, OH, USA
| | - Joshua D Rieskamp
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Elizabeth D Kirby
- Department of Psychology, College of Arts and Sciences, The Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
18
|
Gros A, Furlan FM, Rouglan V, Favereaux A, Bontempi B, Morel JL. Physical exercise restores adult neurogenesis deficits induced by simulated microgravity. NPJ Microgravity 2024; 10:69. [PMID: 38906877 PMCID: PMC11192769 DOI: 10.1038/s41526-024-00411-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
Cognitive impairments have been reported in astronauts during spaceflights and documented in ground-based models of simulated microgravity (SMG) in animals. However, the neuronal causes of these behavioral effects remain largely unknown. We explored whether adult neurogenesis, known to be a crucial plasticity mechanism supporting memory processes, is altered by SMG. Adult male Long-Evans rats were submitted to the hindlimb unloading model of SMG. We studied the proliferation, survival and maturation of newborn cells in the following neurogenic niches: the subventricular zone (SVZ)/olfactory bulb (OB) and the dentate gyrus (DG) of the hippocampus, at different delays following various periods of SMG. SMG exposure for 7 days, but not shorter periods of 6 or 24 h, resulted in a decrease of newborn cell proliferation restricted to the DG. SMG also induced a decrease in short-term (7 days), but not long-term (21 days), survival of newborn cells in the SVZ/OB and DG. Physical exercise, used as a countermeasure, was able to reverse the decrease in newborn cell survival observed in the SVZ and DG. In addition, depending on the duration of SMG periods, transcriptomic analysis revealed modifications in gene expression involved in neurogenesis. These findings highlight the sensitivity of adult neurogenesis to gravitational environmental factors during a transient period, suggesting that there is a period of adaptation of physiological systems to this new environment.
Collapse
Affiliation(s)
- Alexandra Gros
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
- Centre National d'Etudes Spatiales, F-75001, Paris, France
| | - Fandilla Marie Furlan
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
- Department of Genetics & Evolution, 30 Quai Ernest-Ansermet, 1205, Geneva, Switzerland
| | - Vanessa Rouglan
- CNRS, IINS, UMR 5297, University Bordeaux, F-33000, Bordeaux, France
| | | | - Bruno Bontempi
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
| | - Jean-Luc Morel
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France.
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France.
| |
Collapse
|
19
|
Suda K, Pignatelli J, Genis L, Fernandez AM, de Sevilla EF, de la Cruz IF, Pozo-Rodrigalvarez A, de Ceballos ML, Díaz-Pacheco S, Herrero-Labrador R, Aleman IT. A role for astrocytic insulin-like growth factor I receptors in the response to ischemic insult. J Cereb Blood Flow Metab 2024; 44:970-984. [PMID: 38017004 PMCID: PMC11318401 DOI: 10.1177/0271678x231217669] [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: 03/20/2023] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 11/30/2023]
Abstract
Increased neurotrophic support, including insulin-like growth factor I (IGF-I), is an important aspect of the adaptive response to ischemic insult. However, recent findings indicate that the IGF-I receptor (IGF-IR) in neurons plays a detrimental role in the response to stroke. Thus, we investigated the role of astrocytic IGF-IR on ischemic insults using tamoxifen-regulated Cre deletion of IGF-IR in glial fibrillary acidic protein (GFAP) astrocytes, a major cellular component in the response to injury. Ablation of IGF-IR in astrocytes (GFAP-IGF-IR KO mice) resulted in larger ischemic lesions, greater blood-brain-barrier disruption and more deteriorated sensorimotor coordination. RNAseq detected increases in inflammatory, cell adhesion and angiogenic pathways, while the expression of various classical biomarkers of response to ischemic lesion were significantly increased at the lesion site compared to control littermates. While serum IGF-I levels after injury were decreased in both control and GFAP-IR KO mice, brain IGF-I mRNA expression show larger increases in the latter. Further, greater damage was also accompanied by altered glial reactivity as reflected by changes in the morphology of GFAP astrocytes, and relative abundance of ionized calcium binding adaptor molecule 1 (Iba 1) microglia. These results suggest a protective role for astrocytic IGF-IR in the response to ischemic injury.
Collapse
Affiliation(s)
- Kentaro Suda
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jaime Pignatelli
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Laura Genis
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Ana M Fernandez
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | | | | | | | - Maria L de Ceballos
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Sonia Díaz-Pacheco
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Raquel Herrero-Labrador
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Ignacio Torres Aleman
- CIBERNED, Madrid, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
20
|
Balbim GM, Boa Sorte Silva NC, Ten Brinke L, Falck RS, Hortobágyi T, Granacher U, Erickson KI, Hernández-Gamboa R, Liu-Ambrose T. Aerobic exercise training effects on hippocampal volume in healthy older individuals: a meta-analysis of randomized controlled trials. GeroScience 2024; 46:2755-2764. [PMID: 37943486 PMCID: PMC10828456 DOI: 10.1007/s11357-023-00971-7] [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: 06/25/2023] [Accepted: 10/04/2023] [Indexed: 11/10/2023] Open
Abstract
We conducted a meta-analysis of randomized controlled trials investigating the effects of aerobic exercise training (AET) lasting ≥ 4 weeks on hippocampal volume and cardiorespiratory fitness (CRF) in cognitively unimpaired, healthy older individuals. Random-effects robust variance estimation models were used to test differences between AET and controls, while meta-regressions tested associations between CRF and hippocampal volume changes. We included eight studies (N = 554) delivering fully supervised AET for 3 to 12 months (M = 7.8, SD = 4.5) with an average AET volume of 129.85 min/week (SD = 45.5) at moderate-to-vigorous intensity. There were no significant effects of AET on hippocampal volume (SMD = 0.10, 95% CI - 0.01 to 0.21, p = 0.073), but AET moderately improved CRF (SMD = 0.30, 95% CI 0.12 to 0.48, p = 0.005). Improvement in CRF was not associated with changes in hippocampal volume (bSE = 0.05, SE = 0.51, p = 0.923). From the limited number of studies, AET does not seem to impact hippocampal volume in cognitively unimpaired, healthy older individuals. Notable methodological limitations across investigations might mask the lack of effects.
Collapse
Affiliation(s)
- Guilherme Moraes Balbim
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Nárlon Cássio Boa Sorte Silva
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Lisanne Ten Brinke
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Ryan S Falck
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Tibor Hortobágyi
- Center for Human Movement Sciences, University of Groningen Medical Center, Groningen, the Netherlands
- Department of Kinesiology, Hungarian University of Sports Science, Budapest, Hungary
- Department of Sport Biology, Institute of Sport Sciences and Physical Education, University of Pécs, Pécs, Hungary
- Department of Neurology, Somogy County Kaposi Mór Teaching Hospital, Kaposvár, Hungary
| | - Urs Granacher
- Department of Sport and Sport Science, Exercise and Human Movement Science, University of Freiburg, Freiburg, Germany
| | - Kirk I Erickson
- AdventHealth Research Institute, Neuroscience, Orlando, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, USA
| | - Rebeca Hernández-Gamboa
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
- Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, Canada.
| |
Collapse
|
21
|
Tsuruya K, Yoshida H. Cognitive Impairment and Brain Atrophy in Patients with Chronic Kidney Disease. J Clin Med 2024; 13:1401. [PMID: 38592226 PMCID: PMC10931800 DOI: 10.3390/jcm13051401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 04/10/2024] Open
Abstract
In Japan, the aging of the population is rapidly accelerating, with an increase in patients with chronic kidney disease (CKD) and those undergoing dialysis. As a result, the number of individuals with cognitive impairment (CI) is rising, and addressing this issue has become an urgent problem. A notable feature of dementia in CKD patients is the high frequency of vascular dementia, making its prevention through the management of classical risk factors such as hypertension, diabetes mellitus, dyslipidemia, smoking, etc., associated with atherosclerosis and arteriosclerosis. Other effective measures, including the use of renin-angiotensin system inhibitors, addressing anemia, exercise therapy, and lifestyle improvements, have been reported. The incidence and progression of CI may also be influenced by the type of kidney replacement therapy, with reports suggesting that long-duration dialysis, low-temperature hemodialysis, peritoneal dialysis, and kidney transplantation can have a preferable effect on the preservation of cognitive function. In conclusion, patients with CKD are at a higher risk of developing CI, with brain atrophy being a contributing factor. Despite the identification of various preventive measures, the evidence substantiating their efficacy remains limited across all studies. Future expectations lie in large-scale randomized controlled trials.
Collapse
Affiliation(s)
- Kazuhiko Tsuruya
- Department of Nephrology, Nara Medical University, Kashihara 634-8521, Nara, Japan
| | - Hisako Yoshida
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Osaka, Japan;
| |
Collapse
|
22
|
Fong H, Zhou B, Feng H, Luo C, Bai B, Zhang J, Wang Y. Recapitulation of Structure-Function-Regulation of Blood-Brain Barrier under (Patho)Physiological Conditions. Cells 2024; 13:260. [PMID: 38334652 PMCID: PMC10854731 DOI: 10.3390/cells13030260] [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: 10/30/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
The blood-brain barrier (BBB) is a remarkable and intricate barrier that controls the exchange of molecules between the bloodstream and the brain. Its role in maintaining the stability of the central nervous system cannot be overstated. Over the years, advancements in neuroscience and technology have enabled us to delve into the cellular and molecular components of the BBB, as well as its regulation. Yet, there is a scarcity of comprehensive reviews that follow a logical framework of structure-function-regulation, particularly focusing on the nuances of BBB regulation under both normal and pathological conditions. This review sets out to address this gap by taking a historical perspective on the discovery of the BBB and highlighting the major observations that led to its recognition as a distinct brain barrier. It explores the intricate cellular elements contributing to the formation of the BBB, including endothelial cells, pericytes, astrocytes, and neurons, emphasizing their collective role in upholding the integrity and functionality of the BBB. Furthermore, the review delves into the dynamic regulation of the BBB in physiological states, encompassing neural, humoral, and auto-regulatory mechanisms. By shedding light on these regulatory processes, a deeper understanding of the BBB's response to various physiological cues emerges. This review also investigates the disruption of the BBB integrity under diverse pathological conditions, such as ischemia, infection, and toxin exposure. It elucidates the underlying mechanisms that contribute to BBB dysfunction and explores potential therapeutic strategies that aim to restore the BBB integrity and function. Overall, this recapitulation provides valuable insights into the structure, functions, and regulation of the BBB. By integrating historical perspectives, cellular elements, regulatory mechanisms, and pathological implications, this review contributes to a more comprehensive understanding of the BBB and paves the way for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Hin Fong
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - Botao Zhou
- Department of Physiology, Basic Medical and Public Health School, Jinan University, Guangzhou 510632, China;
| | - Haixiao Feng
- Gies College of Business, University of Illinois Urbana-Champaign, Urbana-Champaign, IL 61801, USA;
| | - Chuoying Luo
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - Boren Bai
- Faculty of Medicine, International School, Jinan University, Guangzhou 510632, China; (H.F.); (C.L.); (B.B.)
| | - John Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92350, USA;
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuechun Wang
- Department of Physiology, Basic Medical and Public Health School, Jinan University, Guangzhou 510632, China;
| |
Collapse
|
23
|
Chen R, Zhao B, Huang J, Zhang M, Wang Y, Fu J, Liang H, Zhan H. The Effects of Different Exercise Interventions on Patients with Subjective Cognitive Decline: A Systematic Review and Network Meta-Analysis. J Prev Alzheimers Dis 2024; 11:620-631. [PMID: 38706278 PMCID: PMC11060994 DOI: 10.14283/jpad.2024.65] [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: 11/28/2023] [Accepted: 02/18/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND AND OBJECTIVE Exercise is a promising non-pharmacological therapy for subjective cognitive decline, but it is unclear which type of exercise is most effective. The objective was to assess the comparative effects and ranks of all exercise-based interventions on cognitive function in patients with subjective cognitive decline (SCD). METHOD In this network meta-analysis, Online databases for Web of Science, PubMed, Embase, Medline, Cochrane Library and PsycINFO were searched from inception to April 30, 2023. The included studies are randomized controlled trials assessing the efficacy of exercise interventions for individuals with SCD. The primary outcome measure is memory, while secondary outcome measures encompass executive function, attention, verbal fluency, and global cognitive function. Represented using Standardized Mean Differences (SMDs) along with their 95% Confidence Intervals (CIs). Bias assessment was conducted in accordance with the 'Cochrane Risk of Bias Assessment Tool, 2nd Edition' (RoB 2). Pairwise meta-analysis was carried out using the 'meta-analysis' module within STATA 14.0, and network meta-analysis was performed using the 'mvmeta' and 'network' packages available in STATA 14.0. Registration number CRD42023289687. RESULT This study included a total of 11 randomized controlled trials, encompassing 1,166 patients. Mind-body exercise was found to be efficacious in enhancing or sustaining memory (SMD: 0.58, 95%CI: 0.06 ~ 1.10) and executive function (SMD: 0.41, 95%CI: 0.09 ~ 0.73) in individuals with subjective cognitive decline. Furthermore, mind-body exercise exhibited the highest probability of being the most effective measures for improving or preventing the decline in memory (surface under cumulative ranking curve (SUCRA) value: 90.4) and executive function (SUCRA value: 91.8). The second-ranked moderate-intensity aerobic exercise has also shown a positive effect on the improvement of executive function in patients with subjective cognitive decline (SMD: 0.23, 95%CI: 0.03 ~ 0.43, SUCRA value: 68.2). However, we did not observe a significant effectiveness of exercise interventions on verbal fluency, attention, and overall cognitive function in subjective cognitive decline. CONCLUSION Mind-body exercise may potentially be the optimal strategies for enhancing memory and executive function in individuals with subjective cognitive decline. Additionally, moderate-intensity aerobic exercise has shown a modest positive effect on executive function in subjective cognitive decline. When resources permit, practical application of these findings may be considered. Nevertheless, further support for the conclusions of this study is warranted through larger sample sizes and well-designed multicenter trials.
Collapse
Affiliation(s)
- R Chen
- Hongrui Zhan, Zhuhai, Guangdong, China. Tel: +86 13823017727. E-mail address:
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Findlay MC, Kundu M, Nelson JR, Cole KL, Winterton C, Tenhoeve S, Lucke-Wold B. Emerging Treatments for Subarachnoid Hemorrhage. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1345-1356. [PMID: 38409689 DOI: 10.2174/0118715273279212240130065713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/19/2023] [Accepted: 01/01/2024] [Indexed: 02/28/2024]
Abstract
The current landscape of therapeutic strategies for subarachnoid hemorrhage (SAH), a significant adverse neurological event commonly resulting from the rupture of intracranial aneurysms, is rapidly evolving. Through an in-depth exploration of the natural history of SAH, historical treatment approaches, and emerging management modalities, the present work aims to provide a broad overview of the shifting paradigms in SAH care. By synthesizing the historical management protocols with contemporary therapeutic advancements, patient-specific treatment plans can be individualized and optimized to deliver outstanding care for the best possible SAH-related outcomes.
Collapse
Affiliation(s)
- Matthew C Findlay
- Department of Neurosurgery, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM Hospital, Bhubaneswar, India
| | - Jayson R Nelson
- Department of Neurosurgery, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Kyril L Cole
- Department of Neurosurgery, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Candace Winterton
- Department of Neurosurgery, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Samuel Tenhoeve
- Department of Neurosurgery, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
25
|
Talbot JS, Perkins DR, Tallon CM, Dawkins TG, Douglas AJM, Beckerleg R, Crofts A, Wright ME, Davies S, Steventon JJ, Murphy K, Lord RN, Pugh CJA, Oliver JL, Lloyd RS, Ainslie PN, McManus AM, Stembridge M. Cerebral blood flow and cerebrovascular reactivity are modified by maturational stage and exercise training status during youth. Exp Physiol 2023; 108:1500-1515. [PMID: 37742137 PMCID: PMC10988468 DOI: 10.1113/ep091279] [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: 05/04/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023]
Abstract
NEW FINDINGS What is the central question of this study? Gonadal hormones modulate cerebrovascular function while insulin-like growth factor 1 (IGF-1) facilitates exercise-mediated cerebral angiogenesis; puberty is a critical period of neurodevelopment alongside elevated gonadal hormone and IGF-1 activity: but whether exercise training across puberty enhances cerebrovascular function is unkown. What is the main finding and its importance? Cerebral blood flow is elevated in endurance trained adolescent males when compared to untrained counterparts. However, cerebrovascular reactivity to hypercapnia is faster in trained vs. untrained children, but not adolescents. Exercise-induced improvements in cerebrovascular function are attainable as early as the first decade of life. ABSTRACT Global cerebral blood flow (gCBF) and cerebrovascular reactivity to hypercapnia (CV R C O 2 ${\mathrm{CV}}{{\mathrm{R}}_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ ) are modulated by gonadal hormone activity, while insulin-like growth factor 1 facilitates exercise-mediated cerebral angiogenesis in adults. Whether critical periods of heightened hormonal and neural development during puberty represent an opportunity to further enhance gCBF andCV R C O 2 ${\mathrm{CV}}{{\mathrm{R}}_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ is currently unknown. Therefore, we used duplex ultrasound to assess gCBF andCV R C O 2 ${\mathrm{CV}}{{\mathrm{R}}_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ in n = 128 adolescents characterised as endurance-exercise trained (males: n = 30, females: n = 36) or untrained (males: n = 29, females: n = 33). Participants were further categorised as pre- (males: n = 35, females: n = 33) or post- (males: n = 24, females: n = 36) peak height velocity (PHV) to determine pubertal or 'maturity' status. Three-factor ANOVA was used to identify main and interaction effects of maturity status, biological sex and training status on gCBF andCV R C O 2 ${\mathrm{CV}}{{\mathrm{R}}_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ . Data are reported as group means (SD). Pre-PHV youth demonstrated elevated gCBF and slowerCV R C O 2 ${\mathrm{CV}}{{\mathrm{R}}_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ mean response times than post-PHV counterparts (both: P ≤ 0.001). gCBF was only elevated in post-PHV trained males when compared to untrained counterparts (634 (43) vs. 578 (46) ml min-1 ; P = 0.007). However,CV R C O 2 ${\mathrm{CV}}{{\mathrm{R}}_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ mean response time was faster in pre- (72 (20) vs. 95 (29) s; P ≤ 0.001), but not post-PHV (P = 0.721) trained youth when compared to untrained counterparts. Cardiorespiratory fitness was associated with gCBF in post-PHV youth (r2 = 0.19; P ≤ 0.001) andCV R C O 2 ${\mathrm{CV}}{{\mathrm{R}}_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ mean response time in pre-PHV youth (r2 = 0.13; P = 0.014). Higher cardiorespiratory fitness during adolescence can elevate gCBF while exercise training during childhood primes the development of cerebrovascular function, highlighting the importance of exercise training during the early stages of life in shaping the cerebrovascular phenotype.
Collapse
Affiliation(s)
- Jack S. Talbot
- Cardiff School of Sport and Health SciencesCardiff Metropolitan UniversityCardiffUK
- Centre for Health, Activity and Wellbeing ResearchCardiff Metropolitan UniversityCardiffUK
| | - Dean R. Perkins
- Department of Sport ScienceUniversity of InnsbruckInnsbruckAustria
| | - Christine M. Tallon
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise SciencesUniversity of British Columbia OkanaganKelownaCanada
| | - Tony G. Dawkins
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise SciencesUniversity of British Columbia OkanaganKelownaCanada
| | - Andrew J. M. Douglas
- Cardiff School of Sport and Health SciencesCardiff Metropolitan UniversityCardiffUK
- Centre for Health, Activity and Wellbeing ResearchCardiff Metropolitan UniversityCardiffUK
| | - Ryan Beckerleg
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and AstronomyCardiff UniversityCardiffUK
| | - Andrew Crofts
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and AstronomyCardiff UniversityCardiffUK
| | - Melissa E. Wright
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and AstronomyCardiff UniversityCardiffUK
| | - Saajan Davies
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and AstronomyCardiff UniversityCardiffUK
| | - Jessica J. Steventon
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and AstronomyCardiff UniversityCardiffUK
| | - Kevin Murphy
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Physics and AstronomyCardiff UniversityCardiffUK
| | - Rachel N. Lord
- Cardiff School of Sport and Health SciencesCardiff Metropolitan UniversityCardiffUK
- Centre for Health, Activity and Wellbeing ResearchCardiff Metropolitan UniversityCardiffUK
| | - Christopher J. A. Pugh
- Cardiff School of Sport and Health SciencesCardiff Metropolitan UniversityCardiffUK
- Centre for Health, Activity and Wellbeing ResearchCardiff Metropolitan UniversityCardiffUK
| | - Jon L. Oliver
- Youth Physical Development CentreCardiff Metropolitan UniversityCardiffUK
- Sports Performance Research Institute New ZealandAUT UniversityAucklandNew Zealand
| | - Rhodri S. Lloyd
- Youth Physical Development CentreCardiff Metropolitan UniversityCardiffUK
- Sports Performance Research Institute New ZealandAUT UniversityAucklandNew Zealand
- Centre for Sport Science and Human PerformanceWaikato Institute of TechnologyWaikatoNew Zealand
| | - Philip N. Ainslie
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise SciencesUniversity of British Columbia OkanaganKelownaCanada
| | - Ali M. McManus
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise SciencesUniversity of British Columbia OkanaganKelownaCanada
| | - Mike Stembridge
- Cardiff School of Sport and Health SciencesCardiff Metropolitan UniversityCardiffUK
- Centre for Health, Activity and Wellbeing ResearchCardiff Metropolitan UniversityCardiffUK
- Youth Physical Development CentreCardiff Metropolitan UniversityCardiffUK
| |
Collapse
|
26
|
Silvin A, Qian J, Ginhoux F. Brain macrophage development, diversity and dysregulation in health and disease. Cell Mol Immunol 2023; 20:1277-1289. [PMID: 37365324 PMCID: PMC10616292 DOI: 10.1038/s41423-023-01053-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Brain macrophages include microglia in the parenchyma, border-associated macrophages in the meningeal-choroid plexus-perivascular space, and monocyte-derived macrophages that infiltrate the brain under various disease conditions. The vast heterogeneity of these cells has been elucidated over the last decade using revolutionary multiomics technologies. As such, we can now start to define these various macrophage populations according to their ontogeny and their diverse functional programs during brain development, homeostasis and disease pathogenesis. In this review, we first outline the critical roles played by brain macrophages during development and healthy aging. We then discuss how brain macrophages might undergo reprogramming and contribute to neurodegenerative disorders, autoimmune diseases, and glioma. Finally, we speculate about the most recent and ongoing discoveries that are prompting translational attempts to leverage brain macrophages as prognostic markers or therapeutic targets for diseases that affect the brain.
Collapse
Affiliation(s)
- Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, 94800, France
| | - Jiawen Qian
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, 94800, France.
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, 138648, Republic of Singapore.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, 169856, Singapore.
| |
Collapse
|
27
|
Cefis M, Chaney R, Wirtz J, Méloux A, Quirié A, Leger C, Prigent-Tessier A, Garnier P. Molecular mechanisms underlying physical exercise-induced brain BDNF overproduction. Front Mol Neurosci 2023; 16:1275924. [PMID: 37868812 PMCID: PMC10585026 DOI: 10.3389/fnmol.2023.1275924] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Accumulating evidence supports that physical exercise (EX) is the most effective non-pharmacological strategy to improve brain health. EX prevents cognitive decline associated with age and decreases the risk of developing neurodegenerative diseases and psychiatric disorders. These positive effects of EX can be attributed to an increase in neurogenesis and neuroplastic processes, leading to learning and memory improvement. At the molecular level, there is a solid consensus to involve the neurotrophin brain-derived neurotrophic factor (BDNF) as the crucial molecule for positive EX effects on the brain. However, even though EX incontestably leads to beneficial processes through BDNF expression, cellular sources and molecular mechanisms underlying EX-induced cerebral BDNF overproduction are still being elucidated. In this context, the present review offers a summary of the different molecular mechanisms involved in brain's response to EX, with a specific focus on BDNF. It aims to provide a cohesive overview of the three main mechanisms leading to EX-induced brain BDNF production: the neuronal-dependent overexpression, the elevation of cerebral blood flow (hemodynamic hypothesis), and the exerkine signaling emanating from peripheral tissues (humoral response). By shedding light on these intricate pathways, this review seeks to contribute to the ongoing elucidation of the relationship between EX and cerebral BDNF expression, offering valuable insights into the potential therapeutic implications for brain health enhancement.
Collapse
Affiliation(s)
- Marina Cefis
- Département des Sciences de l’Activité Physique, Faculté des Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Remi Chaney
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Julien Wirtz
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Alexandre Méloux
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Clémence Leger
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
- Département Génie Biologique, Institut Universitaire de Technologie, Dijon, France
| |
Collapse
|
28
|
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
|
29
|
Zalouli V, Rajavand H, Bayat M, Khaleghnia J, Sharifianjazi F, Jafarinazhad F, Beheshtizadeh N. Adult hippocampal neurogenesis (AHN) controls central nervous system and promotes peripheral nervous system regeneration via physical exercise. Biomed Pharmacother 2023; 165:115078. [PMID: 37390707 DOI: 10.1016/j.biopha.2023.115078] [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: 05/08/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/02/2023] Open
Abstract
Physical exercise has beneficial effects on adult hippocampal neurogenesis (AHN) and cognitive processes, including learning. Although it is not known if anaerobic resistance training and high-intensity interval training, which involve alternating brief bouts of highly intense anaerobic activity with rest periods, have comparable effects on AHN. Also, while less thoroughly investigated, individual genetic diversity in the overall response to physical activity is likely to play a key role in the effects of exercise on AHN. Physical exercise has been shown to improve health on average, although the benefits may vary from person to person, perhaps due to genetic differences. Maximal aerobic capacity and metabolic health may improve significantly with aerobic exercise for some people, while the same amount of training may have little effect on others. This review discusses the AHN's capability for peripheral nervous system (PNS) regeneration and central nervous system (CNS) control via physical exercise. Exercise neurogenicity, effective genes, growth factors, and the neurotrophic factors involved in PNS regeneration and CNS control were discussed. Also, some disorders that could be affected by AHN and physical exercise are summarized.
Collapse
Affiliation(s)
- Vahideh Zalouli
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hosnieh Rajavand
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahdi Bayat
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Medicine and Surgery, Physical Activity and Health Promotion, University of Tor Vergata, Rome, Italy
| | - Jalil Khaleghnia
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Sport Sciences, Khavaran Institute of Higher Education, Mashhad, Iran
| | - Fariborz Sharifianjazi
- Department of Natural Sciences, School of Science and Technology, University of Georgia, Tbilisi 0171, Georgia
| | - Farzad Jafarinazhad
- Yeditepe University, Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Istanbul, Turkey.
| | - Nima Beheshtizadeh
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Talbot JS, Perkins DR, Dawkins TG, Douglas AJM, Griffiths TD, Richards CT, Owen K, Lord RN, Pugh CJA, Oliver JL, Lloyd RS, Ainslie PN, McManus AM, Stembridge M. Neurovascular coupling and cerebrovascular hemodynamics are modified by exercise training status at different stages of maturation during youth. Am J Physiol Heart Circ Physiol 2023; 325:H510-H521. [PMID: 37450291 PMCID: PMC10538977 DOI: 10.1152/ajpheart.00302.2023] [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: 05/23/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Neurovascular coupling (NVC) is mediated via nitric oxide signaling, which is independently influenced by sex hormones and exercise training. Whether exercise training differentially modifies NVC pre- versus postpuberty, where levels of circulating sex hormones will differ greatly within and between sexes, remains to be determined. Therefore, we investigated the influence of exercise training status on resting intracranial hemodynamics and NVC at different stages of maturation. Posterior and middle cerebral artery velocities (PCAv and MCAv) and pulsatility index (PCAPI and MCAPI) were assessed via transcranial Doppler ultrasound at rest and during visual NVC stimuli. N = 121 exercise-trained (males, n = 32; females, n = 32) and untrained (males, n = 28; females, n = 29) participants were characterized as pre (males, n = 33; females, n = 29)- or post (males, n = 27; females, n = 32)-peak height velocity (PHV). Exercise-trained youth demonstrated higher resting MCAv (P = 0.010). Maturity and training status did not affect the ΔPCAv and ΔMCAv during NVC. However, pre-PHV untrained males (19.4 ± 13.5 vs. 6.8 ± 6.0%; P ≤ 0.001) and females (19.3 ± 10.8 vs. 6.4 ± 7.1%; P ≤ 0.001) had a higher ΔPCAPI during NVC than post-PHV untrained counterparts, whereas the ΔPCAPI was similar in pre- and post-PHV trained youth. Pre-PHV untrained males (19.4 ± 13.5 vs. 7.9 ± 6.0%; P ≤ 0.001) and females (19.3 ± 10.8 vs. 11.1 ± 7.3%; P = 0.016) also had a larger ΔPCAPI than their pre-PHV trained counterparts during NVC, but the ΔPCAPI was similar in trained and untrained post-PHV youth. Collectively, our data indicate that exercise training elevates regional cerebral blood velocities during youth, but training-mediated adaptations in NVC are only attainable during early stages of adolescence. Therefore, childhood provides a unique opportunity for exercise-mediated adaptations in NVC.NEW & NOTEWORTHY We report that the change in cerebral blood velocity during a neurovascular coupling task (NVC) is similar in pre- and postpubertal youth, regardless of exercise-training status. However, prepubertal untrained youth demonstrated a greater increase in cerebral blood pulsatility during the NVC task when compared with their trained counterparts. Our findings highlight that childhood represents a unique opportunity for exercise-mediated adaptations in cerebrovascular hemodynamics during NVC, which may confer long-term benefits in cerebrovascular function.
Collapse
Affiliation(s)
- Jack S Talbot
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Centre for Health, Activity and Wellbeing Research, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Dean R Perkins
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Tony G Dawkins
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Andrew J M Douglas
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Centre for Health, Activity and Wellbeing Research, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Thomas D Griffiths
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Centre for Health, Activity and Wellbeing Research, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Cory T Richards
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Centre for Health, Activity and Wellbeing Research, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Kerry Owen
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Windsor Clive Primary School, Cardiff, United Kingdom
| | - Rachel N Lord
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Centre for Health, Activity and Wellbeing Research, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Christopher J A Pugh
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Centre for Health, Activity and Wellbeing Research, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Jon L Oliver
- Youth Physical Development Centre, Cardiff Metropolitan University, Cardiff, United Kingdom
- Sports Performance Research Institute New Zealand, AUT University, Auckland, New Zealand
| | - Rhodri S Lloyd
- Youth Physical Development Centre, Cardiff Metropolitan University, Cardiff, United Kingdom
- Sports Performance Research Institute New Zealand, AUT University, Auckland, New Zealand
- Centre for Sport Science and Human Performance, Waikato Institute of Technology, Waikato, New Zealand
| | - Philip N Ainslie
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Ali M McManus
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Mike Stembridge
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Centre for Health, Activity and Wellbeing Research, Cardiff Metropolitan University, Cardiff, United Kingdom
| |
Collapse
|
31
|
Jiménez Peinado P, Urbach A. From Youthful Vigor to Aging Decline: Unravelling the Intrinsic and Extrinsic Determinants of Hippocampal Neural Stem Cell Aging. Cells 2023; 12:2086. [PMID: 37626896 PMCID: PMC10453598 DOI: 10.3390/cells12162086] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Since Joseph Altman published his pioneering work demonstrating neurogenesis in the hippocampus of adult rats, the number of publications in this field increased exponentially. Today, we know that the adult hippocampus harbors a pool of adult neural stem cells (NSCs) that are the source of life-long neurogenesis and plasticity. The functions of these NSCs are regulated by extrinsic cues arising from neighboring cells and the systemic environment. However, this tight regulation is subject to imbalance with age, resulting in a decline in adult NSCs and neurogenesis, which contributes to the progressive deterioration of hippocampus-related cognitive functions. Despite extensive investigation, the mechanisms underlying this age-related decline in neurogenesis are only incompletely understood, but appear to include an increase in NSC quiescence, changes in differentiation patterns, and NSC exhaustion. In this review, we summarize recent work that has improved our knowledge of hippocampal NSC aging, focusing on NSC-intrinsic mechanisms as well as cellular and molecular changes in the niche and systemic environment that might be involved in the age-related decline in NSC functions. Additionally, we identify future directions that may advance our understanding of NSC aging and the concomitant loss of hippocampal neurogenesis and plasticity.
Collapse
Affiliation(s)
| | - Anja Urbach
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
- Jena Center for Healthy Aging, Jena University Hospital, 07747 Jena, Germany
- Aging Research Center Jena, Leibniz Institute on Aging, 07745 Jena, Germany
| |
Collapse
|
32
|
Nuñez A, Zegarra-Valdivia J, Fernandez de Sevilla D, Pignatelli J, Torres Aleman I. The neurobiology of insulin-like growth factor I: From neuroprotection to modulation of brain states. Mol Psychiatry 2023; 28:3220-3230. [PMID: 37353586 DOI: 10.1038/s41380-023-02136-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/25/2023]
Abstract
After decades of research in the neurobiology of IGF-I, its role as a prototypical neurotrophic factor is undisputed. However, many of its actions in the adult brain indicate that this growth factor is not only involved in brain development or in the response to injury. Following a three-layer assessment of its role in the central nervous system, we consider that at the cellular level, IGF-I is indeed a bona fide neurotrophic factor, modulating along ontogeny the generation and function of all the major types of brain cells, contributing to sculpt brain architecture and adaptive responses to damage. At the circuit level, IGF-I modulates neuronal excitability and synaptic plasticity at multiple sites, whereas at the system level, IGF-I intervenes in energy allocation, proteostasis, circadian cycles, mood, and cognition. Local and peripheral sources of brain IGF-I input contribute to a spatially restricted, compartmentalized, and timed modulation of brain activity. To better define these variety of actions, we consider IGF-I a modulator of brain states. This definition aims to reconcile all aspects of IGF-I neurobiology, and may provide a new conceptual framework in the design of future research on the actions of this multitasking neuromodulator in the brain.
Collapse
Affiliation(s)
- A Nuñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - J Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED, Madrid, Spain
- Universidad Señor de Sipán, Chiclayo, Perú
| | - D Fernandez de Sevilla
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - J Pignatelli
- CIBERNED, Madrid, Spain
- Cajal Institute (CSIC), Madrid, Spain
| | - I Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- CIBERNED, Madrid, Spain.
- Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
33
|
Liu C, Wu X, Vulugundam G, Gokulnath P, Li G, Xiao J. Exercise Promotes Tissue Regeneration: Mechanisms Involved and Therapeutic Scope. SPORTS MEDICINE - OPEN 2023; 9:27. [PMID: 37149504 PMCID: PMC10164224 DOI: 10.1186/s40798-023-00573-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Exercise has well-recognized beneficial effects on the whole body. Previous studies suggest that exercise could promote tissue regeneration and repair in various organs. In this review, we have summarized the major effects of exercise on tissue regeneration primarily mediated by stem cells and progenitor cells in skeletal muscle, nervous system, and vascular system. The protective function of exercise-induced stem cell activation under pathological conditions and aging in different organs have also been discussed in detail. Moreover, we have described the primary molecular mechanisms involved in exercise-induced tissue regeneration, including the roles of growth factors, signaling pathways, oxidative stress, metabolic factors, and non-coding RNAs. We have also summarized therapeutic approaches that target crucial signaling pathways and molecules responsible for exercise-induced tissue regeneration, such as IGF1, PI3K, and microRNAs. Collectively, the comprehensive understanding of exercise-induced tissue regeneration will facilitate the discovery of novel drug targets and therapeutic strategies.
Collapse
Affiliation(s)
- Chang Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Xinying Wu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | | | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
34
|
Lee CJ, Lee HY, Yu YS, Ryu KB, Lee H, Kim K, Shin SY, Gil YC, Cho SJ. Brain compartmentalization based on transcriptome analyses and its gene expression in Octopus minor. Brain Struct Funct 2023:10.1007/s00429-023-02647-6. [PMID: 37138199 DOI: 10.1007/s00429-023-02647-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/17/2023] [Indexed: 05/05/2023]
Abstract
Coleoid cephalopods have a high intelligence, complex structures, and large brain. The cephalopod brain is divided into supraesophageal mass, subesophageal mass and optic lobe. Although much is known about the structural organization and connections of various lobes of octopus brain, there are few studies on the brain of cephalopod at the molecular level. In this study, we demonstrated the structure of an adult Octopus minor brain by histomorphological analyses. Through visualization of neuronal and proliferation markers, we found that adult neurogenesis occurred in the vL and posterior svL. We also obtained specific 1015 genes by transcriptome of O. minor brain and selected OLFM3, NPY, GnRH, and GDF8 genes. The expression of genes in the central brain showed the possibility of using NPY and GDF8 as molecular marker of compartmentation in the central brain. This study will provide useful information for establishing a molecular atlas of cephalopod brain.
Collapse
Affiliation(s)
- Chan-Jun Lee
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Hae-Youn Lee
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Yun-Sang Yu
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Kyoung-Bin Ryu
- Clinical Research Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chungbuk, 28159, Republic of Korea
| | - Hyerim Lee
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Kyunghwan Kim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Song Yub Shin
- Department of Cellular and Molecular Medicine, School of Medicine, Chosun University, Gwangju, 61452, Republic of Korea.
| | - Young-Chun Gil
- Department of Anatomy, College of Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Sung-Jin Cho
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
35
|
Reddy I, Yadav Y, Dey CS. Cellular and Molecular Regulation of Exercise-A Neuronal Perspective. Cell Mol Neurobiol 2023; 43:1551-1571. [PMID: 35986789 PMCID: PMC11412429 DOI: 10.1007/s10571-022-01272-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
The beneficial effects of exercise on the proper functioning of the body have been firmly established. Multi-systemic metabolic regulation of exercise is the consequence of multitudinous changes that occur at the cellular level. The exercise responsome comprises all molecular entities including exerkines, miRNA species, growth factors, signaling proteins that are elevated and activated by physical exercise. Exerkines are secretory molecules released by organs such as skeletal muscle, adipose tissue, liver, and gut as a function of acute/chronic exercise. Exerkines such as FNDC5/irisin, Cathepsin B, Adiponectin, and IL-6 circulate through the bloodstream, cross the blood-brain barrier, and modulate the expression of important signaling molecules such as AMPK, SIRT1, PGC1α, BDNF, IGF-1, and VEGF which further contribute to improved energy metabolism, glucose homeostasis, insulin sensitivity, neurogenesis, synaptic plasticity, and overall well-being of the body and brain. These molecules are also responsible for neuroprotective adaptations that exercise confers on the brain and potentially ameliorate neurodegeneration. This review aims to detail important cellular and molecular species that directly or indirectly mediate exercise-induced benefits in the body, with an emphasis on the central nervous system.
Collapse
Affiliation(s)
- Ishitha Reddy
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Yamini Yadav
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India.
| |
Collapse
|
36
|
Wu MY, Zou WJ, Lee D, Mei L, Xiong WC. APP in the Neuromuscular Junction for the Development of Sarcopenia and Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24097809. [PMID: 37175515 PMCID: PMC10178513 DOI: 10.3390/ijms24097809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/18/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Sarcopenia, an illness condition usually characterized by a loss of skeletal muscle mass and muscle strength or function, is often associated with neurodegenerative diseases, such as Alzheimer's disease (AD), a common type of dementia, leading to memory loss and other cognitive impairment. However, the underlying mechanisms for their associations and relationships are less well understood. The App, a Mendelian gene for early-onset AD, encodes amyloid precursor protein (APP), a transmembrane protein enriched at both the neuromuscular junction (NMJ) and synapses in the central nervous system (CNS). Here, in this review, we highlight APP and its family members' physiological functions and Swedish mutant APP (APPswe)'s pathological roles in muscles and NMJ. Understanding APP's pathophysiological functions in muscles and NMJ is likely to uncover insights not only into neuromuscular diseases but also AD. We summarize key findings from the burgeoning literature, which may open new avenues to investigate the link between muscle cells and brain cells in the development and progression of AD and sarcopenia.
Collapse
Affiliation(s)
- Min-Yi Wu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wen-Jun Zou
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Northeast Ohio VA Healthcare System, Cleveland, OH 44106, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Northeast Ohio VA Healthcare System, Cleveland, OH 44106, USA
| |
Collapse
|
37
|
Karakatsani A, Álvarez-Vergara MI, de Almodóvar CR. The vasculature of neurogenic niches: Properties and function. Cells Dev 2023; 174:203841. [PMID: 37060947 DOI: 10.1016/j.cdev.2023.203841] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
In the adult rodent brain, neural stem cells (NSCs) reside in the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the hippocampus. In these areas, NSCs and their progeny integrate intrinsic signals and extrinsic cues provided by their microenvironment that control their behavior. The vasculature in the SVZ and SGZ, and the choroid plexus (ChP) in the SVZ, have emerged as critical compartments of the neurogenic niches as they provide a rich repertoire of cues to regulate NSC quiescence, proliferation, self-renewal and differentiation. Physical contact between NSCs and blood vessels is also a feature within the niches and supports different processes such as quiescence, migration and vesicle transport. In this review, we provide a description of the brain and choroid plexus vasculature in both stem cell niches, highlighting the main properties and role of the vasculature in each niche. We also summarize the current understanding of how blood vessel- and ChP-derived signals influence the behavior of NSCs in physiological adulthood, as well as upon aging.
Collapse
Affiliation(s)
- Andromachi Karakatsani
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - María I Álvarez-Vergara
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Carmen Ruiz de Almodóvar
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, University of Bonn, Bonn, Germany; Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
38
|
Lee KY, Rhodes JS, Saif MTA. Astrocyte-mediated Transduction of Muscle Fiber Contractions Synchronizes Hippocampal Neuronal Network Development. Neuroscience 2023; 515:25-36. [PMID: 36736611 PMCID: PMC10023357 DOI: 10.1016/j.neuroscience.2023.01.028] [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: 10/08/2022] [Revised: 01/08/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Exercise supports brain health in part by enhancing hippocampal function. The leading hypothesis is that muscles release factors when they contract (e.g., lactate, myokines, growth factors) that enter circulation and reach the brain where they enhance plasticity (e.g., increase neurogenesis and synaptogenesis). However, it remains unknown how the muscle signals are transduced by the hippocampal cells to modulate network activity and synaptic development. Thus, we established an in vitro model in which the media from contracting primary muscle cells (CM) is applied to developing primary hippocampal cell cultures on a microelectrode array. We found that the hippocampal neuronal network matures more rapidly (as indicated by synapse development and synchronous neuronal activity) when exposed to CM than regular media (RM). This was accompanied by a 4.4- and 1.4-fold increase in the proliferation of astrocytes and neurons, respectively. Further, experiments established that factors released by astrocytes inhibit neuronal hyper-excitability induced by muscle media, and facilitate network development. Results provide new insight into how exercise may support hippocampal function by regulating astrocyte proliferation and subsequent taming of neuronal activity into an integrated network.
Collapse
Affiliation(s)
- Ki Yun Lee
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Justin S Rhodes
- Department of Psychology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - M Taher A Saif
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
39
|
Physical activity for cognitive health promotion: An overview of the underlying neurobiological mechanisms. Ageing Res Rev 2023; 86:101868. [PMID: 36736379 DOI: 10.1016/j.arr.2023.101868] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/13/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Physical activity is one of the modifiable factors of cognitive decline and dementia with the strongest evidence. Although many influential reviews have illustrated the neurobiological mechanisms of the cognitive benefits of physical activity, none of them have linked the neurobiological mechanisms to normal exercise physiology to help the readers gain a more advanced, comprehensive understanding of the phenomenon. In this review, we address this issue and provide a synthesis of the literature by focusing on five most studied neurobiological mechanisms. We show that the body's adaptations to enhance exercise performance also benefit the brain and contribute to improved cognition. Specifically, these adaptations include, 1), the release of growth factors that are essential for the development and growth of neurons and for neurogenesis and angiogenesis, 2), the production of lactate that provides energy to the brain and is involved in the synthesis of glutamate and the maintenance of long-term potentiation, 3), the release of anti-inflammatory cytokines that reduce neuroinflammation, 4), the increase in mitochondrial biogenesis and antioxidant enzyme activity that reduce oxidative stress, and 5), the release of neurotransmitters such as dopamine and 5-HT that regulate neurogenesis and modulate cognition. We also discussed several issues relevant for prescribing physical activity, including what intensity and mode of physical activity brings the most cognitive benefits, based on their influence on the above five neurobiological mechanisms. We hope this review helps readers gain a general understanding of the state-of-the-art knowledge on the neurobiological mechanisms of the cognitive benefits of physical activity and guide them in designing new studies to further advance the field.
Collapse
|
40
|
Gao Y, Syed M, Zhao X. Mechanisms underlying the effect of voluntary running on adult hippocampal neurogenesis. Hippocampus 2023; 33:373-390. [PMID: 36892196 PMCID: PMC10566571 DOI: 10.1002/hipo.23520] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/11/2023] [Accepted: 02/17/2023] [Indexed: 03/10/2023]
Abstract
Adult hippocampal neurogenesis is important for preserving learning and memory-related cognitive functions. Physical exercise, especially voluntary running, is one of the strongest stimuli to promote neurogenesis and has beneficial effects on cognitive functions. Voluntary running promotes exit of neural stem cells (NSCs) from the quiescent stage, proliferation of NSCs and progenitors, survival of newborn cells, morphological development of immature neuron, and integration of new neurons into the hippocampal circuitry. However, the detailed mechanisms driving these changes remain unclear. In this review, we will summarize current knowledge with respect to molecular mechanisms underlying voluntary running-induced neurogenesis, highlighting recent genome-wide gene expression analyses. In addition, we will discuss new approaches and future directions for dissecting the complex cellular mechanisms driving change in adult-born new neurons in response to physical exercise.
Collapse
Affiliation(s)
- Yu Gao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Moosa Syed
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
41
|
Liao X, Shen J, Li M. Effects of multi-domain intervention on intrinsic capacity in older adults: A systematic review of randomized controlled trials (RCTs). Exp Gerontol 2023; 174:112112. [PMID: 36736466 DOI: 10.1016/j.exger.2023.112112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Intrinsic capacity is central to the maintenance of function in older adults, and maintaining optimal intrinsic capacity is of great importance to promote healthy aging. The purpose of this systematic review and meta-analysis was to analyze the impact of multi-domain interventions on intrinsic capacity in older adults, intervention components, and potential interactions between components. A total of 6740 published articles were screened until August 2022, and the review included 25 randomized controlled trials that analyzed populations, interventions, control groups, and outcomes. The meta-analysis showed improvements in the primary outcome indicators in the intervention group compared to the control group. These included increased scores on the Mini-Mental State Examination as an indicator of cognitive function, decreased scores on the Geriatric Depression Scale (GDS-15) as an indicator of psychological ability and increased scores on the Short Physical Performance Battery (SPPB) as an indicator of physical performance, with only the SPPB indicator analyzed showing greater heterogeneity. Significant improvements were also seen in the secondary indicators Time-to-Walk Test (TUG), gait speed, Chair Stand Test (CST), grip strength values and BMI. There was insufficient data for the Mini Nutritional Assessment (MNA) as an indicator of vitality to conduct a meta-analysis. Studies were of moderate to high quality. The results of this review indicate that multi-domain interventions can maintain the level of intrinsic capacity in older adults and are equally effective in older adults with declining self-care abilities.
Collapse
Affiliation(s)
- Xiaoyan Liao
- Nursing Department, The First Affiliated Hospital of Chongqing Medical University, Yuzhong district, #1 Youyi road, Chongqing 400014, China
| | - Jun Shen
- Nursing Department, The First Affiliated Hospital of Chongqing Medical University, Yuzhong district, #1 Youyi road, Chongqing 400014, China.
| | - Miao Li
- Nursing Department, The First Affiliated Hospital of Chongqing Medical University, Yuzhong district, #1 Youyi road, Chongqing 400014, China
| |
Collapse
|
42
|
Blood-to-brain communication in aging and rejuvenation. Nat Neurosci 2023; 26:379-393. [PMID: 36646876 DOI: 10.1038/s41593-022-01238-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 11/21/2022] [Indexed: 01/18/2023]
Abstract
Aging induces molecular, cellular and functional changes in the adult brain that drive cognitive decline and increase vulnerability to dementia-related neurodegenerative diseases. Leveraging systemic and lifestyle interventions, such as heterochronic parabiosis, administration of 'young blood', exercise and caloric restriction, has challenged prevalent views of brain aging as a rigid process and has demonstrated that aging-associated cognitive and cellular impairments can be restored to more youthful levels. Technological advances in proteomic and transcriptomic analyses have further facilitated investigations into the functional impact of intertissue communication on brain aging and have led to the identification of a growing number of pro-aging and pro-youthful factors in blood. In this review, we discuss blood-to-brain communication from a systems physiology perspective with an emphasis on blood-derived signals as potent drivers of both age-related brain dysfunction and brain rejuvenation.
Collapse
|
43
|
Graciani AL, Gutierre MU, Coppi AA, Arida RM, Gutierre RC. MYELIN, AGING, AND PHYSICAL EXERCISE. Neurobiol Aging 2023; 127:70-81. [PMID: 37116408 DOI: 10.1016/j.neurobiolaging.2023.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023]
Abstract
Myelin sheath is a structure in neurons fabricated by oligodendrocytes and Schwann cells responsible for increasing the efficiency of neural synapsis, impulse transmission, and providing metabolic support to the axon. They present morpho-functional changes during health aging as deformities of the sheath and its fragmentation, causing an increased load on microglial phagocytosis, with Alzheimer's disease aggravating. Physical exercise has been studied as a possible protective agent for the nervous system, offering benefits to neuroplasticity. In this regard, studies in animal models for Alzheimer's and depression reported the efficiency of physical exercise in protecting against myelin degeneration. A reduction of myelin damage during aging has also been observed in healthy humans. Physical activity promotes oligodendrocyte proliferation and myelin preservation during old age, although some controversies remain. In this review, we will address how effective physical exercise can be as a protective agent of the myelin sheath against the effects of aging in physiological and pathological conditions.
Collapse
|
44
|
Bhattacharya P, Chatterjee S, Roy D. Impact of exercise on brain neurochemicals: a comprehensive review. SPORT SCIENCES FOR HEALTH 2023. [DOI: 10.1007/s11332-022-01030-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
45
|
Arjunan A, Sah DK, Woo M, Song J. Identification of the molecular mechanism of insulin-like growth factor-1 (IGF-1): a promising therapeutic target for neurodegenerative diseases associated with metabolic syndrome. Cell Biosci 2023; 13:16. [PMID: 36691085 PMCID: PMC9872444 DOI: 10.1186/s13578-023-00966-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Neurodegenerative disorders are accompanied by neuronal degeneration and glial dysfunction, resulting in cognitive, psychomotor, and behavioral impairment. Multiple factors including genetic, environmental, metabolic, and oxidant overload contribute to disease progression. Recent evidences suggest that metabolic syndrome is linked to various neurodegenerative diseases. Metabolic syndrome (MetS) is known to be accompanied by symptoms such as hyperglycemia, abdominal obesity, hypertriglyceridemia, and hypertension. Despite advances in knowledge about the pathogenesis of neurodegenerative disorders, effective treatments to combat neurodegenerative disorders caused by MetS have not been developed to date. Insulin growth factor-1 (IGF-1) deficiency has been associated with MetS-related pathologies both in-vivo and in-vitro. IGF-1 is essential for embryonic and adult neurogenesis, neuronal plasticity, neurotropism, angiogenesis, metabolic function, and protein clearance in the brain. Here, we review the evidence for the potential therapeutic effects of IGF-1 in the neurodegeneration related to metabolic syndrome. We elucidate how IGF-1 may be involved in molecular signaling defects that occurs in MetS-related neurodegenerative disorders and highlight the importance of IGF-1 as a potential therapeutic target in MetS-related neurological diseases.
Collapse
Affiliation(s)
- Archana Arjunan
- grid.14005.300000 0001 0356 9399Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-Do 58128 Republic of Korea
| | - Dhiraj Kumar Sah
- grid.14005.300000 0001 0356 9399Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128 Republic of Korea ,grid.14005.300000 0001 0356 9399BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun, 58128 Republic of Korea
| | - Minna Woo
- grid.17063.330000 0001 2157 2938Division of Endocrinology and Metabolism, University Health Network and and Banting and Best Diabetes Centre, University of Toronto, Toronto, ON Canada
| | - Juhyun Song
- grid.14005.300000 0001 0356 9399Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-Do 58128 Republic of Korea ,grid.14005.300000 0001 0356 9399BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 Seoyangro, Hwasun, 58128 Republic of Korea
| |
Collapse
|
46
|
Wooliscroft L, McCoy S, Hildebrand A, Rooney W, Oken BS, Spain RI, Kuehl KS, Bourdette D, Cameron M. Protocol for an exploratory, randomised, single-blind clinical trial of aerobic exercise to promote remyelination in multiple sclerosis. BMJ Open 2023; 13:e061539. [PMID: 36596632 PMCID: PMC9814998 DOI: 10.1136/bmjopen-2022-061539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION There is an urgent need for remyelinating therapies that restore function in people with multiple sclerosis (pwMS). Aerobic exercise is a promising remyelinating strategy because it promotes remyelination in animal models both independently and synergistically with medications. Here, in this study, we present an innovative, randomised, single-blind, clinical trial designed to explore: the relationship between demyelination and mobility (part 1), and if 24 weeks of aerobic exercise promotes remyelination in pwMS (part 2). METHODS AND ANALYSIS Sedentary participants (n=60; aged 18-64 years) with stable MS will undergo a baseline visit with the following outcomes to assess associations between demyelination and mobility (part 1): spinal cord demyelination (somatosensory-evoked potentials, SSEPs), mobility (6-Minute Timed Walk, Timed 25-Foot Walk, Timed Up and Go, 9-Hole Peg Test) and patient-reported outcomes (PROs). After baseline testing, participants with significantly prolonged SSEP latency will advance to the clinical exercise trial (part 2) and will be randomised 1:1 to active or control conditions for 24 weeks. The active condition will be aerobic stationary cycling three times per week with graded virtual supervision. The control condition will be monthly virtual MS symptom education groups (six sessions). SSEP latency (remyelination endpoint), mobility outcomes and PROs will be measured at 12 and 24 weeks in all clinical trial participants. A subset of 11 active and 11 control participants will undergo a brain MRI with quantitative T1 myelin water fraction at baseline and 24 weeks (exploratory remyelination endpoint). ETHICS AND DISSEMINATION Ethical approval was obtained from the Oregon Health & Science University Institutional Review Board (#21045). Dissemination of findings will include peer-reviewed publications, conference presentations and media releases. The proposed study will inform the feasibility, study design and sample size for a fully powered clinical trial of aerobic exercise to promote remyelination in pwMS. TRIAL REGISTRATION NUMBER NCT04539002.
Collapse
Affiliation(s)
- Lindsey Wooliscroft
- Neurology, Oregon Health & Science University, Portland, Oregon, USA
- Neurology, Portland VA Medical Center, Portland, Oregon, USA
| | - Sharon McCoy
- Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Andrea Hildebrand
- Biostatistics and Design Program Core, Oregon Health & Science University, Portland, Oregon, USA
| | - William Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Barry S Oken
- Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Rebecca Irene Spain
- Neurology, Oregon Health & Science University, Portland, Oregon, USA
- Neurology, Portland VA Medical Center, Portland, Oregon, USA
| | - Kerry S Kuehl
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Dennis Bourdette
- Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Michelle Cameron
- Neurology, Oregon Health & Science University, Portland, Oregon, USA
- Neurology, Portland VA Medical Center, Portland, Oregon, USA
| |
Collapse
|
47
|
Colasuonno F, Price R, Moreno S. Upper and Lower Motor Neurons and the Skeletal Muscle: Implication for Amyotrophic Lateral Sclerosis (ALS). ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:111-129. [PMID: 37955773 DOI: 10.1007/978-3-031-38215-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The relationships between motor neurons and the skeletal muscle during development and in pathologic contexts are addressed in this Chapter.We discuss the developmental interplay of muscle and nervous tissue, through neurotrophins and the activation of differentiation and survival pathways. After a brief overview on muscular regulatory factors, we focus on the contribution of muscle to early and late neurodevelopment. Such a role seems especially intriguing in relation to the epigenetic shaping of developing motor neuron fate choices. In this context, emphasis is attributed to factors regulating energy metabolism, which may concomitantly act in muscle and neural cells, being involved in common pathways.We then review the main features of motor neuron diseases, addressing the cellular processes underlying clinical symptoms. The involvement of different muscle-associated neurotrophic factors for survival of lateral motor column neurons, innervating MyoD-dependent limb muscles, and of medial motor column neurons, innervating Myf5-dependent back musculature is discussed. Among the pathogenic mechanisms, we focus on oxidative stress, that represents a common and early trait in several neurodegenerative disorders. The role of organelles primarily involved in reactive oxygen species scavenging and, more generally, in energy metabolism-namely mitochondria and peroxisomes-is discussed in the frame of motor neuron degeneration.We finally address muscular involvement in amyotrophic lateral sclerosis (ALS), a multifactorial degenerative disorder, hallmarked by severe weight loss, caused by imbalanced lipid metabolism. Even though multiple mechanisms have been recognized to play a role in the disease, current literature generally assumes that the primum movens is neuronal degeneration and that muscle atrophy is only a consequence of such pathogenic event. However, several lines of evidence point to the muscle as primarily involved in the disease, mainly through its role in energy homeostasis. Data from different ALS mouse models strongly argue for an early mitochondrial dysfunction in muscle tissue, possibly leading to motor neuron disturbances. Detailed understanding of skeletal muscle contribution to ALS pathogenesis will likely lead to the identification of novel therapeutic strategies.
Collapse
Affiliation(s)
- Fiorella Colasuonno
- Department of Experimental Medicine , University of Rome "Tor Vergata", Rome, Italy
- Department of Science, LIME, University Roma Tre, Rome, Italy
| | - Rachel Price
- Department of Science, LIME, University Roma Tre, Rome, Italy
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Sandra Moreno
- Department of Science, LIME, University Roma Tre, Rome, Italy.
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
48
|
Barnes JN, Burns JM, Bamman MM, Billinger SA, Bodine SC, Booth FW, Brassard P, Clemons TA, Fadel PJ, Geiger PC, Gujral S, Haus JM, Kanoski SE, Miller BF, Morris JK, O’Connell KM, Poole DC, Sandoval DA, Smith JC, Swerdlow RH, Whitehead SN, Vidoni ED, van Praag H. Proceedings from the Albert Charitable Trust Inaugural Workshop on 'Understanding the Acute Effects of Exercise on the Brain'. Brain Plast 2022; 8:153-168. [PMID: 36721393 PMCID: PMC9837736 DOI: 10.3233/bpl-220146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
An inaugural workshop supported by "The Leo and Anne Albert Charitable Trust," was held October 4-7, 2019 in Scottsdale, Arizona, to focus on the effects of exercise on the brain and to discuss how physical activity may prevent or delay the onset of aging-related neurodegenerative conditions. The Scientific Program Committee (led by Dr. Jeff Burns) assembled translational, clinical, and basic scientists who research various aspects of the effects of exercise on the body and brain, with the overall goal of gaining a better understanding as to how to delay or prevent neurodegenerative diseases. In particular, research topics included the links between cardiorespiratory fitness, the cerebrovasculature, energy metabolism, peripheral organs, and cognitive function, which are all highly relevant to understanding the effects of acute and chronic exercise on the brain. The Albert Trust workshop participants addressed these and related topics, as well as how other lifestyle interventions, such as diet, affect age-related cognitive decline associated with Alzheimer's and other neurodegenerative diseases. This report provides a synopsis of the presentations and discussions by the participants, and a delineation of the next steps towards advancing our understanding of the effects of exercise on the aging brain.
Collapse
Affiliation(s)
- Jill N. Barnes
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, USA
| | - Marcas M. Bamman
- UAB Center for Exercise Medicine, University of Alabama, Birmingham, AL, USA
| | | | - Sue C. Bodine
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Frank W. Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Patrice Brassard
- Department of Kinesiology, Faculty of Medicine, Université Laval, and Research center of the Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, QC, Canada
| | - Tameka A. Clemons
- Department of Professional and Medical Education, Meharry Medical College, Nashville, TN, USA
| | - Paul J. Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| | - Paige C. Geiger
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Swathi Gujral
- University of Pittsburgh School of Medicine, Department of Psychiatry, Pittsburgh, PA, USA
| | - Jacob M. Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Scott E. Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsrife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Benjamin F. Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jill K. Morris
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, USA
| | | | - David C. Poole
- Departments of Kinesiology, Anatomy and Physiology, Kansas State University, Manhattan, KS, USA
| | | | - J. Carson Smith
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD, USA
| | | | - Shawn N. Whitehead
- Vulnerable Brain Laboratory, Department Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, N6A 5C1, Canada
| | - Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, USA
| | - Henriette van Praag
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter FL, USA
| |
Collapse
|
49
|
Savignac C, Villeneuve S, Badhwar A, Saltoun K, Shafighi K, Zajner C, Sharma V, Gagliano Taliun SA, Farhan S, Poirier J, Bzdok D. APOE alleles are associated with sex-specific structural differences in brain regions affected in Alzheimer's disease and related dementia. PLoS Biol 2022; 20:e3001863. [PMID: 36512526 PMCID: PMC9747055 DOI: 10.1371/journal.pbio.3001863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/30/2022] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease is marked by intracellular tau aggregates in the medial temporal lobe (MTL) and extracellular amyloid aggregates in the default network (DN). Here, we examined codependent structural variations between the MTL's most vulnerable structure, the hippocampus (HC), and the DN at subregion resolution in individuals with Alzheimer's disease and related dementia (ADRD). By leveraging the power of the approximately 40,000 participants of the UK Biobank cohort, we assessed impacts from the protective APOE ɛ2 and the deleterious APOE ɛ4 Alzheimer's disease alleles on these structural relationships. We demonstrate ɛ2 and ɛ4 genotype effects on the inter-individual expression of HC-DN co-variation structural patterns at the population level. Across these HC-DN signatures, recurrent deviations in the CA1, CA2/3, molecular layer, fornix's fimbria, and their cortical partners related to ADRD risk. Analyses of the rich phenotypic profiles in the UK Biobank cohort further revealed male-specific HC-DN associations with air pollution and female-specific associations with cardiovascular traits. We also showed that APOE ɛ2/2 interacts preferentially with HC-DN co-variation patterns in estimating social lifestyle in males and physical activity in females. Our structural, genetic, and phenotypic analyses in this large epidemiological cohort reinvigorate the often-neglected interplay between APOE ɛ2 dosage and sex and link APOE alleles to inter-individual brain structural differences indicative of ADRD familial risk.
Collapse
Affiliation(s)
- Chloé Savignac
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- Department of Neurology and Neurosurgery, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- McConnell Brain Imaging Centre (BIC), MNI, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Centre for Studies in the Prevention of Alzheimer’s Disease, Douglas Mental Health Institute, McGill University, Montreal, Quebec, Canada
| | - AmanPreet Badhwar
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Centre de recherche de l’Institut universitaire de gériatrie de Montréal (CRIUGM), Montreal, Quebec, Canada
| | - Karin Saltoun
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Kimia Shafighi
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Chris Zajner
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Vaibhav Sharma
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sarah A. Gagliano Taliun
- Department of Neurosciences & Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Montréal, Quebec, Canada
| | - Sali Farhan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Judes Poirier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Centre for Studies in the Prevention of Alzheimer’s Disease, Douglas Mental Health Institute, McGill University, Montreal, Quebec, Canada
| | - Danilo Bzdok
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- McConnell Brain Imaging Centre (BIC), MNI, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- School of Computer Science, McGill University, Montreal, Quebec, Canada
- Mila—Quebec Artificial Intelligence Institute, Montreal, Quebec, Canada
| |
Collapse
|
50
|
Malin SK, Stewart NR, Ude AA, Alderman BL. Brain insulin resistance and cognitive function: influence of exercise. J Appl Physiol (1985) 2022; 133:1368-1380. [PMID: 36269295 PMCID: PMC9744647 DOI: 10.1152/japplphysiol.00375.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
Exercise has systemic health benefits in people, in part, through improving whole body insulin sensitivity. The brain is an insulin-sensitive organ that is often underdiscussed relative to skeletal muscle, liver, and adipose tissue. Although brain insulin action may have only subtle impacts on peripheral regulation of systemic glucose homeostasis, it is important for weight regulation as well as mental health. In fact, brain insulin signaling is also involved in processes that support healthy cognition. Furthermore, brain insulin resistance has been associated with age-related declines in memory and executive function as well as Alzheimer's disease pathology. Herein, we provide an overview of brain insulin sensitivity in relation to cognitive function from animal and human studies, with particular emphasis placed on the impact exercise may have on brain insulin sensitivity. Mechanisms discussed include mitochondrial function, brain growth factors, and neurogenesis, which collectively help combat obesity-related metabolic disease and Alzheimer's dementia.
Collapse
Affiliation(s)
- Steven K Malin
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
- Division of Endocrinology, Metabolism & Nutrition, Rutgers University, New Brunswick, New Jersey
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| | - Nathan R Stewart
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
| | - Andrew A Ude
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
| | - Brandon L Alderman
- Department of Kinesiology & Health, Rutgers University, New Brunswick, New Jersey
- Center of Alcohol and Substance Use Studies, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|