1
|
Shi JX, Wang ZY, Wang SW, Shen Q, Tan X. Exercise-mediated muscle-hypothalamus crosstalk: Improvement for cognitive dysfunction caused by disrupted circadian rhythm. Life Sci 2025; 373:123657. [PMID: 40306358 DOI: 10.1016/j.lfs.2025.123657] [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: 02/18/2025] [Revised: 04/14/2025] [Accepted: 04/20/2025] [Indexed: 05/02/2025]
Abstract
In contemporary societal evolution, the increasing disruption of the natural sleep-wake cycle, attributable to factors such as shift work and overexposure to artificial light, has been paralleled by a marked escalation in the incidence of cognitive impairments and the prevalence of neurodegenerative diseases. Current management strategies for cognitive impairments include pharmacological and non-pharmacological interventions. Pharmacological interventions for cognitive impairments typically involve medications to manage cognitive symptoms and improve neurological functions. However, these drugs show limited long-term efficacy in slowing disease progression and may cause side effects. Given the widespread occurrence of cognitive dysfunction, it is crucial to develop accessible non-pharmacological interventions. Physical activity and exercise have emerged as pivotal lifestyle determinants known to exert a modulatory effect on the risk profile for cognitive dysfunction caused by disrupted circadian rhythms. The skeletal muscle, a dynamic tissue, undergoes a profound morphological and metabolic reconfiguration in response to physical exertion, along with the secretion of myokines. Additionally, the hypothalamus, particularly the ventromedial nuclei, arcuate nuclei, and the suprachiasmatic nucleus, have crucial functions in regulating physical activity, influencing energy metabolism, and managing circadian cycles. Nevertheless, the communication between the hypothalamus and skeletal muscle during exercise is not fully understood. This narrative review integrates current knowledge on the interaction between the hypothalamus and skeletal muscle during exercise, emphasizing its neuroendocrine effects and potential therapeutic implications for alleviating cognitive dysfunction associated with disrupted circadian rhythms.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Zi-Yuan Wang
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Sheng-Wen Wang
- School of Basic Medical Science, Naval Medical University, Shanghai 200433, China
| | - Qi Shen
- Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Xing Tan
- Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| |
Collapse
|
2
|
Kim E, Tanzi RE, Choi SH. Therapeutic potential of exercise-hormone irisin in Alzheimer's disease. Neural Regen Res 2025; 20:1555-1564. [PMID: 38993140 PMCID: PMC11688551 DOI: 10.4103/nrr.nrr-d-24-00098] [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: 01/24/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024] Open
Abstract
Irisin is a myokine that is generated by cleavage of the membrane protein fibronectin type III domain-containing protein 5 (FNDC5) in response to physical exercise. Studies reveal that irisin/FNDC5 has neuroprotective functions against Alzheimer's disease, the most common form of dementia in the elderly, by improving cognitive function and reducing amyloid-β and tau pathologies as well as neuroinflammation in cell culture or animal models of Alzheimer's disease. Although current and ongoing studies on irisin/FNDC5 show promising results, further mechanistic studies are required to clarify its potential as a meaningful therapeutic target for alleviating Alzheimer's disease. We recently found that irisin treatment reduces amyloid-β pathology by increasing the activity/levels of amyloid-β-degrading enzyme neprilysin secreted from astrocytes. Herein, we present an overview of irisin/FNDC5's protective roles and mechanisms against Alzheimer's disease.
Collapse
Affiliation(s)
- Eunhee Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
3
|
Wilson AC, Pountney DL, Khoo TK. Therapeutic Mechanisms of Exercise in Parkinson's Disease. Int J Mol Sci 2025; 26:4860. [PMID: 40429998 PMCID: PMC12112306 DOI: 10.3390/ijms26104860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Despite being the second-most common neurodegenerative disease, the etiology of Parkinson's disease (PD) remains uncertain with current knowledge suggestive of multiple risk factors. Furthermore, curative treatment does not yet exist, and treatment is primarily symptomatic in nature. For this reason, supportive therapies such as exercise are a crucial tool in PD management. It is useful to better understand how exercise affects the brain and body in the context of PD to guide clinical decision-making and determine the optimal exercise intensity and modality for PD patients. This review outlines the various mechanisms by which exercise can be beneficial as a therapeutic option in PD.
Collapse
Affiliation(s)
- Alice C. Wilson
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia
| | - Dean L. Pountney
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia
| | - Tien K. Khoo
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia
- Graduate School of Medicine, University of Wollongong, Wollongong, NSW 2500, Australia
- Northern New South Wales Local Health District, NSW Health, Lismore, NSW 2480, Australia
| |
Collapse
|
4
|
Minuti A, Raffaele I, Scuruchi M, Lui M, Muscarà C, Calabrò M. Role and Functions of Irisin: A Perspective on Recent Developments and Neurodegenerative Diseases. Antioxidants (Basel) 2025; 14:554. [PMID: 40427436 PMCID: PMC12108254 DOI: 10.3390/antiox14050554] [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] [Received: 04/04/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Irisin is a peptide derived from fibronectin type III domain-containing protein 5 (FNDC5) and is primarily produced by muscle fibers under the regulation of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) during exercise. Irisin has been the subject of extensive research due to its potential as a metabolic regulator and its antioxidant properties. Notably, it has been associated with protective actions within the brain. Despite growing interest, many questions remain regarding the molecular mechanisms underlying its effects. This review summarizes recent findings on irisin, highlighting its pleiotropic functions and the biological processes and molecular cascades involved in its action, with a particular focus on the central nervous system. Irisin plays a crucial role in neuron survival, differentiation, growth, and development, while also promoting mitochondrial homeostasis, regulating apoptosis, and facilitating autophagy-processes essential for normal neuronal function. Emerging evidence suggests that irisin may improve conditions associated with non-communicable neurological diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia, and multiple sclerosis. Given its diverse benefits, irisin holds promise as a novel therapeutic agent for preventing and treating neurological diseases.
Collapse
Affiliation(s)
- Aurelio Minuti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (A.M.); (M.C.)
| | - Ivana Raffaele
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (A.M.); (M.C.)
| | - Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy;
| | - Maria Lui
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (A.M.); (M.C.)
| | - Claudia Muscarà
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (A.M.); (M.C.)
| | - Marco Calabrò
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (A.M.); (M.C.)
| |
Collapse
|
5
|
Tsuda T. Preventive effects against cognitive decline and obesity via the combination of dietary factors and exercise: can dietary factors replace or complement the beneficial effects of exercise? Food Funct 2025; 16:3243-3254. [PMID: 40197497 DOI: 10.1039/d4fo06349j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Regular exercise is known to have positive effects on a variety of health outcomes, including the prevention of obesity and cognitive decline. Several recent reports have suggested that combining exercise with dietary factors such as flavonoids can amplify health functions. While the addition of dietary factors synergistically or additively enhances the beneficial effects of exercise, the addition of exercise may also reduce the amount of dietary factors needed to obtain their beneficial effects. This review describes (1) the exercise-induced improvement of health functions focusing on cognitive function, (2) several studies that investigated the effects that combining flavonoids with exercise has on obesity and cognitive function, and (3) the synergistic effect of inducing beige adipocyte formation to potentially explain the mechanism of the preventive effects of combining dietary factors and exercise on obesity, including new findings on lactate obtained through these studies. Finally, we summarize the following challenges and issues in research on developing the combination of dietary factors and exercise: clarifying the appropriate types and intensities of exercise and appropriate intake of dietary factors for obtaining the optimal combined effect, accumulating results of human interventional studies, and examining the benefits of this combination in improving the memory and learning ability of young adults and children. As scientists studying functional foods, we should focus more on exercise-replacement and exercise-complementing dietary factors in our research. This journal will play an important role in elucidating the molecular mechanisms underlying the combined effects of dietary factors and exercise.
Collapse
Affiliation(s)
- Takanori Tsuda
- School of Bioscience and Biotechnology and Graduate School of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi 487-8501, Japan.
| |
Collapse
|
6
|
Zhang QX, Zhang LJ, Zhao N, Yang L. Irisin restrains neuroinflammation in mouse experimental autoimmune encephalomyelitis via regulating microglia activation. Front Pharmacol 2025; 16:1561939. [PMID: 40365310 PMCID: PMC12069398 DOI: 10.3389/fphar.2025.1561939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Multiple sclerosis is a chronic autoimmune demyelinating disorder predominantly affecting the white matter of the central nervous system, with experimental autoimmune encephalomyelitis (EAE) serving as its classical animal model. Irisin, a glycosylated protein derived from the proteolytic cleavage of fibronectin type III domain-containing protein 5, plays a significant role in metabolic regulation and inflammatory modulation within the organism. Methods In this study, we systematically investigated the therapeutic effects and underlying mechanism of Irisin on EAE and BV2 microglial cells through comprehensive methodologies including quantitative real-time polymerase chain reaction, immunofluorescence staining and western blot. Results Irisin exerts neuroprotective effects in EAE mice, significantly ameliorating both clinical and pathological manifestations of the disease. Mechanistically, Irisin attenuated inflammatory response and reduced the number of microglia through NF-κBp65 signaling pathway. Conclusion In conclusion, these results collectively suggest that Irisin alleviates EAE progression by suppressing microglia activation via the NF-κBp65 pathway, highlighting its potential as a promising therapeutic target for multiple sclerosis treatment.
Collapse
Affiliation(s)
| | | | | | - Li Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
7
|
Capasso G, Franssen CFM, Perna AF, Massy ZA, Menzies RI, Zoccali C, Tessitore A, Nedergaard M, Okusa MD, Ortiz A, Wagner CA, Unwin RJ. Drivers and mechanisms of cognitive decline in chronic kidney disease. Nat Rev Nephrol 2025:10.1038/s41581-025-00963-0. [PMID: 40281076 DOI: 10.1038/s41581-025-00963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
Cognitive impairment is highly prevalent among individuals with chronic kidney disease (CKD). Despite its high prevalence, the contributing factors and mechanisms underlying brain-kidney dysfunction in CKD remain poorly understood. However, advances in neuroscience, including novel imaging techniques and cognitive assessment methods, have begun to clarify this complex relationship. Several factors contribute directly to cognitive decline in people with CKD, including accumulation of uraemic toxins, microvascular damage, malnutrition, chronic inflammation and disruptions in key neuroprotective pathways, such as those involving Klotho and the glymphatic system. These factors are also linked to the accelerated ageing observed in people with CKD, a key contributor to cognitive decline. However, most studies on cognition in people with CKD have been cross-sectional and associative, offering limited insight into causation. Research advances, such as studies on the effect of uraemic toxins on the blood-brain barrier and the role of the endothelial glycocalyx in vascular damage, offer promising new directions. Emerging data from longitudinal cohort studies are also enhancing our understanding of these processes, with potential implications for both the treatment of CKD-related cognitive decline and the broader issue of cognitive dysfunction in ageing populations. Here, we examine key mechanisms linking CKD to cognitive decline and consider potential therapeutic interventions.
Collapse
Affiliation(s)
- Giovambattista Capasso
- Biogem, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy.
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy.
| | - Casper F M Franssen
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alessandra F Perna
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ziad A Massy
- AURA (Association pour l'Utilization du Rein Artificiel dans la Region Parisienne) Paris, Department of Nephrology, CHU Ambroise Paré, AP-HP, Paris, France
- Centre for Research in Epidemiology and Population Health (CESP), University Paris-Saclay, University Versailles-Saint Quentin, Inserm UMRS 1018, Clinical Epidemiology Team, Villejuif, France
| | - Robert I Menzies
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Carmine Zoccali
- Biogem, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio, Italy
| | - Alessandro Tessitore
- Department of Advanced Surgical and Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maiken Nedergaard
- Center for Basic and Translational Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mark D Okusa
- Division of Nephrology, Center for Immunity Inflammation and Regenerative Medicine University of Virginia, Charlottesville, VA, USA
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD UAM), Madrid, Spain
| | - Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center University of Zurich, Zurich, Switzerland
| | - Robert J Unwin
- UCL Centre for Kidney and Bladder Health, University College London, London, UK
| |
Collapse
|
8
|
Lin H, Yin L, Liu W, Li R, Jiang T, Yang M, Cao Y, Wang S, Yu Y, Chen C, Guo X, Wang W, Liu H, Dai Y, Yan J, Lin Y, Ding Y, Ruan C, Yang L, Wu T, Tao J, Chen L. Muscle-Derived Small Extracellular Vesicles Mediate Exercise-Induced Cognitive Protection in Chronic Cerebral Hypoperfusion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2410209. [PMID: 40271743 DOI: 10.1002/advs.202410209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 04/06/2025] [Indexed: 04/25/2025]
Abstract
Physical exercise protects against cognitive impairment caused by chronic cerebral hypoperfusion (CCH). However, the mechanisms through which exercise sends signals from the periphery to the central nervous system remain incompletely understood. This study demonstrated that exercise promotes the secretion of muscle-derived small extracellular vesicles (sEVs), which facilitate interorgan communication between the muscle and the brain. Systematic delivery of muscle-derived sEVs enhances synaptic plasticity and alleviated cognitive impairment in CCH. Notably, miRNA sequencing reveal miR-17/20a-5p as key cargos in sEVs involved in the exercise-induced muscle-brain crosstalk. Muscle-derived sEVs are also identified as the primary source of swimming-induced miR-17/20a-5p in circulating sEVs. Mechanistically, miR-17/20a-5p binds to the DEP-domain containing mTOR-interacting protein (DEPTOR) and activates the mammalian target of rapamycin (mTOR) pathway in the hippocampus. Depletion of miR-17/20a-5p from muscle-derived sEVs impairs the exercise-induced enhancement of synaptic plasticity and cognitive function. Moreover, overexpression of DEPTOR in the hippocampus attenuates the cognitive benefits of exercise. Conversely, hippocampus-specific activation of mTOR reverses these effects, highlighting the crucial role of mTOR in mediating the positive effects of exercise. Collectively, these findings identify miR-17/20a-5p in muscle-derived sEVs as the exercise-induced myokine with potent effects on the brain, emphasizing the therapeutic potential of exercise in managing cognitive impairment.
Collapse
Affiliation(s)
- Huawei Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Lianhua Yin
- The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350003, China
| | - Weilin Liu
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Rui Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Tao Jiang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Minguang Yang
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yajun Cao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Sinuo Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yan Yu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Cong Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Xiaoqin Guo
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Wenju Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Huanhuan Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yaling Dai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Jiamin Yan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yanting Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Yanyi Ding
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| | - Chendong Ruan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Lei Yang
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Tiecheng Wu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Lidan Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Provincial and Ministerial Co-founded Collaborative Innovation Center of Rehabilitation Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
- Key Laboratory of Cognitive Rehabilitation of Fujian Province, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China
| |
Collapse
|
9
|
Curovic I. The role of resistance exercise-induced local metabolic stress in mediating systemic health and functional adaptations: could condensed training volume unlock greater benefits beyond time efficiency? Front Physiol 2025; 16:1549609. [PMID: 40313877 PMCID: PMC12045103 DOI: 10.3389/fphys.2025.1549609] [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: 12/21/2024] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
The majority of "specialised" exercise configurations (e.g., supersets, drop sets, blood flow restriction) are being assessed as "shortcuts" to hypertrophy and strength improvements. However, these advanced training techniques may also offer significant benefits for systemic health and functional outcomes across recreational and clinical populations via locally induced metabolic responses. Stress-regulating mechanisms are known to enhance the body's resilience by facilitating allostasis, the process of coordinating adaptive processes in reaction to stressors such as physical training. Yet, the role of the local metabolic stress provoked by resistance exercise has not gained much research attention despite its wide potential. Positive effects are not only linked to improved muscular endurance, hypertrophy and strength via primary and secondary mechanisms, but also to the release of myokines, hormones, microRNAs, immune factors, inflammatory substances and other endocrine molecules that initiate numerous health-promoting modifications on a systemic level. Resistance exercise strategies that maximise the local accumulation of metabolites are not well defined, although high volume, close proximity to failure and shorter rests seem to be a necessity. Additionally, blood flow restriction training provides a potent alternative for inducing local acidosis, thereby triggering several pathways associated with improved immunity and physical function even in remote muscle tissues. Future research is warranted to further explore advanced resistance training techniques, as these approaches may offer comparable benefits for physical and mental health to those seen with other forms of exercise such as high-intensity interval training and heavy resistance training.
Collapse
Affiliation(s)
- Ivan Curovic
- Institute of Coaching and Performance, University of Central Lancashire, Preston, United Kingdom
| |
Collapse
|
10
|
Ha J, Sung S, Kim H. Myokines and interorgan crosstalk: bridging exercise to health promotion and disease prevention. Ann Pediatr Endocrinol Metab 2025; 30:59-68. [PMID: 40335041 PMCID: PMC12061759 DOI: 10.6065/apem.2448218.109] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 05/09/2025] Open
Abstract
Exercise is known to promote physical health and reduce the risk of various diseases. During exercise, skeletal muscle actively contracts to perform movements and secretes hormone-like molecules termed myokines. The beneficial effects of exercise have been assessed with respect to myokine production, and those of irisin on bone, adipose tissue, and the brain have been well documented. Irisin, through its interactions with the integrin αV family, plays a crucial role in bone maintenance, metabolic regulation, and cognitive function. Building on the established understanding of irisin, this discussion will examine the functions and effects of other myokines as key secretory factors in exercise, emphasizing their broader roles in health promotion and the potential for new therapeutic strategies in disease prevention and treatment.
Collapse
Affiliation(s)
- Junseo Ha
- Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Suchan Sung
- Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Hyeonwoo Kim
- Korea Advanced Institute of Science and Technology, Daejeon, Korea
| |
Collapse
|
11
|
Taha HB, Birnbaum A, Matthews I, Aceituno K, Leon J, Thorwald M, Godoy-Lugo J, Cortes CJ. Activation of the muscle-to-brain axis ameliorates neurocognitive deficits in an Alzheimer's disease mouse model via enhancing neurotrophic and synaptic signaling. GeroScience 2025; 47:1593-1613. [PMID: 39269584 PMCID: PMC11978596 DOI: 10.1007/s11357-024-01345-3] [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: 07/02/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Skeletal muscle regulates central nervous system (CNS) function and health, activating the muscle-to-brain axis through the secretion of skeletal muscle-originating factors ("myokines") with neuroprotective properties. However, the precise mechanisms underlying these benefits in the context of Alzheimer's disease (AD) remain poorly understood. To investigate muscle-to-brain axis signaling in response to amyloid β (Aβ)-induced toxicity, we generated 5xFAD transgenic female mice with enhanced skeletal muscle function (5xFAD;cTFEB;HSACre) at prodromal (4-months old) and late (8-months old) symptomatic stages. Skeletal muscle TFEB overexpression reduced Aβ plaque accumulation in the cortex and hippocampus at both ages and rescued behavioral neurocognitive deficits in 8-month-old 5xFAD mice. These changes were associated with transcriptional and protein remodeling of neurotrophic signaling and synaptic integrity, partially due to the CNS-targeting myokine prosaposin (PSAP). Our findings implicate the muscle-to-brain axis as a novel neuroprotective pathway against amyloid pathogenesis in AD.
Collapse
Affiliation(s)
- Hash Brown Taha
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Allison Birnbaum
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Ian Matthews
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Karel Aceituno
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Jocelyne Leon
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Max Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Jose Godoy-Lugo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Constanza J Cortes
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA.
| |
Collapse
|
12
|
Wagner CA, Massy ZA, Capasso G, Mattace-Raso F, Pepin M, Bobot M, Zoccali C, Ferreira AC, Hoorn EJ, Imenez Silva PH, Unwin RJ, Pesic V. Translational research on cognitive impairment in chronic kidney disease. Nephrol Dial Transplant 2025; 40:621-631. [PMID: 39400744 DOI: 10.1093/ndt/gfae229] [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: 07/27/2024] [Indexed: 10/15/2024] Open
Abstract
Cognitive decline is common in patients with acute or chronic kidney disease. Several areas of brain function can be affected, including short- and long-term memory, attention and inhibitory control, sleep, mood, eating control and motor function. Cognitive decline in kidney disease shares risk factors with cognitive dysfunction in people without kidney disease, such as diabetes, high blood pressure, sedentary lifestyle and unhealthy diet. However, additional kidney-specific risk factors may contribute, such as uremic toxins, electrolyte imbalances, chronic inflammation, acid-base disorders or endocrine dysregulation. Traditional and kidney-specific risk factors may interact to cause damage to the blood-brain barrier, induce vascular damage in the brain and cause neurotoxicity or neuroinflammation. Here, we discuss recent insights into the pathomechanisms of cognitive decline from animal models and novel avenues for prevention and therapy. We focus on a several areas that influence cognition: blood-brain barrier disruption, the role of skeletal muscle, physical activity and the endocrine factor irisin, and the emerging therapeutic role of sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 (GLP-1) receptor agonists. Taken together, these studies demonstrate the importance of animal models in providing a mechanistic understanding of this complex condition and their potential to explain the mechanisms of novel therapies.
Collapse
Affiliation(s)
- Carsten A Wagner
- Department of Physiology and Zurich Kidney Center (ZKC), University of Zurich, Zurich, Switzerland
| | - Ziad A Massy
- Clinical Epidemiology, Inserm Unit 1018, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS) Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA), Paris, France and Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt/Paris, France
| | - Giovambattista Capasso
- Biogem, Research Institute for Biology and Molecular Genetics, Ariano Irpino, Italy
- Department of Translational Medical Sciences, University of Campania, Luigi Vanvitelli, Napoli, Italy
| | - Francesco Mattace-Raso
- Department of Internal Medicine, Division of Geriatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marion Pepin
- Clinical Epidemiology, Inserm Unit 1018, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS) Villejuif, France
- Geriatric Department, Ambroise Paré University Hospital, APHP, Versailles Saint-Quentin-en-Yvelines University (UVSQ), Boulogne-Billancourt, France
| | - Mickaël Bobot
- Aix-Marseille Univ, C2VN, INSERM 1263, INRAE 1260, CERIMED, Marseille, France
| | - Carmine Zoccali
- Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale, Grande Ospedale Metropolitano, c/o Nefrologia, Reggio Calabria, Italy
| | - Ana C Ferreira
- Unidade Local de Saúde de São José - Hospital Curry Cabral, Nephrology Department, Lisbon, Portugal
- Nova Medical School - Nephrology, Lisbon, Portugal
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Hypertension, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Pedro H Imenez Silva
- Department of Internal Medicine, Division of Nephrology and Hypertension, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert J Unwin
- Department of Renal Medicine, Royal Free Hospital Trust, University College London (UCL), London, UK
| | - Vesna Pesic
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
13
|
Barros-Aragão FGQ, Januszkiewicz E, Hunter T, Lyra E Silva NDM, De Felice FG. Physical activity in Alzheimer's disease prevention: Sex differences and the roles of BDNF and irisin. Front Neuroendocrinol 2025; 77:101189. [PMID: 40228745 DOI: 10.1016/j.yfrne.2025.101189] [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: 11/14/2024] [Revised: 03/07/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD) disproportionately affects women, with postmenopausal hormonal changes contributing to elevated risk. Physical exercise is a promising, non-pharmacological strategy to mitigate cognitive decline and AD progression. Brain-derived neurotrophic factor (BDNF) and irisin are key molecular mediators of exercise-induced brain health and protection against AD pathology by promoting synaptic plasticity, neurogenesis, and reducing amyloidosis, tau pathology, and neuroinflammation in sex-specific mechanisms. This review explores sex and gender influences on exercise outcomes and their interaction with FNDC5/irisin and BDNF signaling pathways in the context of AD prevention. We highlight emerging evidence on the interplay between exercise, sex, and neuroprotective pathways, emphasizing the need for sex-sensitive research designs to advance precision approaches for AD prevention.
Collapse
Affiliation(s)
- F G Q Barros-Aragão
- Centre for Neuroscience Studies, Department for Biomedical and Molecular Sciences, Queen's University, Kingston, Canada; D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
| | - E Januszkiewicz
- Centre for Neuroscience Studies, Department for Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - T Hunter
- Centre for Neuroscience Studies, Department for Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - N de M Lyra E Silva
- Centre for Neuroscience Studies, Department for Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - F G De Felice
- Centre for Neuroscience Studies, Department for Biomedical and Molecular Sciences, Queen's University, Kingston, Canada; D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Bayfield J, Elford HR, Christie BR. Examining a role for irisin in treating cerebral ischemia. J Neurophysiol 2025; 133:1320-1328. [PMID: 40135689 DOI: 10.1152/jn.00027.2025] [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: 01/21/2025] [Revised: 02/09/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
Stroke is a leading cause of death and disability, with ischemic stroke representing most cases. Age is the most significant nonmodifiable risk factor for stroke, and with an aging population, there is an urgent need for effective prevention and treatment strategies. Physical inactivity is a strong risk factor for stroke, and exercise has long been held as a promising approach to improve poststroke outcomes. During exercise, the myokine irisin is released as a product of a type 1 membrane protein cleavage that is encoded by the fibronectin type III domain containing 5 (FNDC5) gene. This review summarizes recent literature on irisin's role in ischemic stroke, examining central effects, stroke risk, poststroke functional outcomes, and exogenous administration. Irisin has value as a prognostic marker for risk stratification. Low levels of irisin correlate with worse outcomes and higher mortality in patients with ischemic stroke. Irisin may also be a key to the benefits of exercise, particularly for high-intensity resistance training, which significantly increases irisin levels. Beyond exercise, exogenous irisin is neuroprotective in murine models, reducing brain edema, inflammation, and apoptosis, and increasing blood-brain barrier integrity and brain-derived neurotrophic factor levels. This underscores irisin's potential to mitigate ischemic damage and promote recovery. Human trials are necessary to validate these findings and explore the feasibility of irisin-based interventions in acute stroke care. This review lays a foundation for future research to clarify irisin's therapeutic benefits, establish optimal exercise protocols, and explore exogenous irisin as a novel intervention for ischemic stroke.
Collapse
Affiliation(s)
- Jack Bayfield
- Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hanna R Elford
- International Collaboration on Repair Discoveries, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian R Christie
- Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
- Department of Psychology, San Diego State University, San Diego, California, United States
| |
Collapse
|
15
|
Sprenger HG, Mittenbühler MJ, Sun Y, Van Vranken JG, Schindler S, Jayaraj A, Khetarpal SA, Smythers AL, Vargas-Castillo A, Puszynska AM, Spinelli JB, Armani A, Kunchok T, Ryback B, Seo HS, Song K, Sebastian L, O'Young C, Braithwaite C, Dhe-Paganon S, Burger N, Mills EL, Gygi SP, Paulo JA, Arthanari H, Chouchani ET, Sabatini DM, Spiegelman BM. Ergothioneine controls mitochondrial function and exercise performance via direct activation of MPST. Cell Metab 2025; 37:857-869.e9. [PMID: 39965563 DOI: 10.1016/j.cmet.2025.01.024] [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: 04/05/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/20/2025]
Abstract
Ergothioneine (EGT) is a diet-derived, atypical amino acid that accumulates to high levels in human tissues. Reduced EGT levels have been linked to age-related disorders, including neurodegenerative and cardiovascular diseases, while EGT supplementation is protective in a broad range of disease and aging models. Despite these promising data, the direct and physiologically relevant molecular target of EGT has remained elusive. Here, we use a systematic approach to identify how mitochondria remodel their metabolome in response to exercise training. From these data, we find that EGT accumulates in muscle mitochondria upon exercise training. Proteome-wide thermal stability studies identify 3-mercaptopyruvate sulfurtransferase (MPST) as a direct molecular target of EGT; EGT binds to and activates MPST, thereby boosting mitochondrial respiration and exercise training performance in mice. Together, these data identify the first physiologically relevant EGT target and establish the EGT-MPST axis as a molecular mechanism for regulating mitochondrial function and exercise performance.
Collapse
Affiliation(s)
- Hans-Georg Sprenger
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Melanie J Mittenbühler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yizhi Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sebastian Schindler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Abhilash Jayaraj
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Sumeet A Khetarpal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Amanda L Smythers
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ariana Vargas-Castillo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Anna M Puszynska
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jessica B Spinelli
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrea Armani
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tenzin Kunchok
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Birgitta Ryback
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Kijun Song
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Luke Sebastian
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Coby O'Young
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Nils Burger
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Evanna L Mills
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Haribabu Arthanari
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - David M Sabatini
- Institute of Organic Chemistry and Biochemistry, Prague, Czech Republic
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Welc SS, Brotto M, White KE, Bonewald LF. Aging: A struggle for beneficial to overcome negative factors made by muscle and bone. Mech Ageing Dev 2025; 224:112039. [PMID: 39952614 PMCID: PMC11893237 DOI: 10.1016/j.mad.2025.112039] [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: 10/28/2024] [Revised: 12/15/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Musculoskeletal health is strongly influenced by regulatory interactions of bone and muscle. Recent discoveries have identified a number of key mechanisms through which soluble factors released during exercise by bone exert positive effects on muscle and by muscle on bone. Although exercise can delay the negative effects of aging, these beneficial effects are diminished with aging. The limited response of aged muscle and bone tissue to exercise are accompanied by a failure in bone and muscle communication. Here, we propose that exercise induced beneficial factors must battle changes in circulating endocrine and inflammatory factors that occur with aging. Furthermore, sedentary behavior results in the release of negative factors impacting the ability of bone and muscle to respond to physical activity especially with aging. In this review we report on exercise responsive factors and evidence of modification occurring with aging.
Collapse
Affiliation(s)
- Steven S Welc
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas-Arlington, Arlington, TX 76019, USA.
| | - Kenneth E White
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Molecular and Medical Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Lynda F Bonewald
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
17
|
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
|
18
|
Wang J, Gao S, Fu S, Li Y, Su L, Li X, Wu G, Jiang J, Zhao Z, Yang C, Wang X, Cui K, Sun X, Qi X, Wang C, Sun H, Shao S, Tian Y, Gong T, Luo J, Zheng J, Cui S, Liao F, Liu F, Wang D, Wong CCL, Yi M, Wan Y. Irisin reprograms microglia through activation of STAT6 and prevents cognitive dysfunction after surgery in mice. Brain Behav Immun 2025; 125:68-91. [PMID: 39701329 DOI: 10.1016/j.bbi.2024.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is common in the aged population and associated with poor clinical outcomes. Irisin, an endogenous molecule that mediates the beneficial effects of exercise, has shown neuroprotective potential in several models of neurological diseases. Here we show that preoperative serum level of irisin is reduced in dementia patients over the age of 70. Comprehensive proteomics analysis reveals that deletion of irisin affects the nervous and immune systems, and reduces the expression of complement proteins. Systemically administered irisin penetrates the blood-brain barrier in mice, targets the microglial integrin αVβ5 receptor, activates signal transducer and activator of transcription 6 (STAT6), induces microglia reprogramming to the M2 phenotype, and improves immune microenvironment in LPS-induced neuroinflammatory mice. Finally, prophylactic administration of irisin prevents POCD-like behavior, particularly early cognitive dysfunction. Our findings provide new insights into the direct regulation of the immune microenvironment by irisin, and reveal that recombinant irisin holds great promise as a novel therapy for preventing POCD and other neuroinflammatory disorders. SUMMARY: Our findings reveal molecular and cellular mechanisms of irisin on neuroinflammation, and show that prophylactic administration of irisin prevents POCD-like behavior, particularly early cognitive dysfunction.
Collapse
Affiliation(s)
- Jiaxin Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Shuaixin Gao
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China; Human Nutrition Program, Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Su Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Yawei Li
- Department of Anesthesiology, Peking University First Hospital, Beijing 10034, China
| | - Li Su
- Peking University Medical and Health Analysis Center, Peking University, Beijing 10034, China
| | - Xiaoman Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Guanghao Wu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jiankuo Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Zifang Zhao
- Department of Pain Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Chaojuan Yang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Xiaoyi Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Kun Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Beijing Life Science Academy, Beijing 102209, China
| | - Xiaoyan Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Xuetao Qi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Cheng Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Changping Laboratory, Beijing 102206, China
| | - Haojie Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Shan Shao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Yue Tian
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Tingting Gong
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Jianyuan Luo
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Shuang Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Feifei Liao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China.
| | - Dongxin Wang
- Department of Anesthesiology, Peking University First Hospital, Beijing 10034, China.
| | - Catherine C L Wong
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China.
| | - Ming Yi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Medical Innovation Center (Taizhou) of Peking University, Taizhou 225316, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Key Laboratory for Neuroscience, Ministry of Education and National Health Commission, Peking University, Beijing, 100191, China; Medical Innovation Center (Taizhou) of Peking University, Taizhou 225316, China.
| |
Collapse
|
19
|
Booranasuksakul U, Guan Z, Macdonald IA, Tsintzas K, Stephan BCM, Siervo M. Sarcopenic obesity and brain health: A critical appraisal of the current evidence. NUTR BULL 2025; 50:30-43. [PMID: 39799465 DOI: 10.1111/nbu.12725] [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: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 01/15/2025]
Abstract
Sarcopenic obesity (SO) is a body composition phenotype derived from the simultaneous presence in the same individual of an increase in fat mass and a decrease in skeletal muscle mass and/or function. Several protocols for the diagnosis of SO have been proposed in the last two decades making prevalence and disease risk estimates of SO heterogeneous and challenging to interpret. Dementia is a complex neurological disorder that significantly impacts patients, carers and healthcare systems. The identification of risk factors for early cognitive impairment and dementia is key to mitigating the forecasted trends of a 2-fold increase in dementia case numbers over the next two decades worldwide. Excess adiposity and sarcopenia have both been independently associated with risk of cognitive impairment and dementia. Whether SO is associated with a greater risk of cognitive impairment and dementia is currently uncertain. This review critically appraises the current evidence on the association between SO with cognitive outcomes and dementia risk. It also discusses some of the putative biological mechanisms that may link the SO phenotype with alteration of brain functions.
Collapse
Affiliation(s)
- Uraiporn Booranasuksakul
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Zhongyang Guan
- Curtin School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, WA, Australia
| | - Ian A Macdonald
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Kostas Tsintzas
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Blossom C M Stephan
- Dementia Centre of Excellence, enAble Institute, Curtin University, Bentley, WA, Australia
| | - Mario Siervo
- Curtin School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, WA, Australia
- Dementia Centre of Excellence, enAble Institute, Curtin University, Bentley, WA, Australia
| |
Collapse
|
20
|
Zhang QH, Jin LM, Lin MS, Wang MX, Cui YQ, Ye JX, Xiong YQ, Luo W, Zhu WW, Liang G. FNDC5/Irisin exacerbates APAP-induced acute liver injury through activating JNK/NF-κB and inflammatory response. Acta Pharmacol Sin 2025:10.1038/s41401-025-01509-7. [PMID: 40016523 DOI: 10.1038/s41401-025-01509-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025]
Abstract
Acute liver injury (ALI) is associated with high mortality rates. Despite its severity, there are currently no effective interventions, underscoring the urgent need for research on the mechanisms driving ALI progression. Irisin, a hormone derived from its precursor FNDC5, has been shown to play a critical role in some chronic liver diseases. In this study we investigated the role of hepatic FNDC5/Irisin in a mouse model of AILI induced by acetaminophen (APAP, 400 mg/kg, i.p.). The mice were euthanized at 6, 12 and 24 h after APAP injection, then the blood and liver tissues were collected for analyses. By conducting transcriptome sequencing, we identified that both the expression and release of FNDC5/Irisin were significantly increased and highly correlated with AILI. We showed that knockout of Irisin significantly improved APAP-induced tissue damage and hepatocyte death in mouse liver. Conversely, preinjection of recombinant Irisin protein (1 mg·kg-1·d-1, i.p., for 3 days) exacerbated the AILI in FNDC5 knockout mice. RNA-seq analysis revealed that knockout of FNDC5/Irisin reduced inflammatory responses and JNK/NF-κB activation in APAP-treated mouse liver, while exogenous Irisin administration aggravated JNK/NF-κB-mediated inflammation. In primary mouse hepatocytes treated with APAP (15 mM), application of Irisin (100 ng/mL) activated the integrin αV/JNK/NF-κB axis, driving inflammation and oxidative stress. In summary, this study highlights Irisin as a critical regulator in AILI progression. Circulating Irisin could be a novel biomarker for AILI diagnosis, and targeting FNDC5/Irisin could hold promise for the development of novel treatments for AILI.
Collapse
Affiliation(s)
- Qian-Hui Zhang
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lei-Ming Jin
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Meng-Sha Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Min-Xiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ya-Qian Cui
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jia-Xi Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yong-Qiang Xiong
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Wu Luo
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- The Affiliated Cangnan Hospital, Wenzhou Medical University, Wenzhou, 325800, China.
| | - Wei-Wei Zhu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- The Affiliated Cangnan Hospital, Wenzhou Medical University, Wenzhou, 325800, China.
| | - Guang Liang
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
21
|
Wang S, Xu H, Liu G, Chen L. Non-pharmacological treatment of Alzheimer's disease: an update. Front Aging Neurosci 2025; 17:1527242. [PMID: 40018518 PMCID: PMC11865074 DOI: 10.3389/fnagi.2025.1527242] [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: 11/13/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that significantly impairs memory, cognitive function, and the ability to perform daily tasks. The pathological features of AD include β-amyloid plaques, neurofibrillary tangles, and neuronal loss. Current AD treatments target pathological changes but often fail to noticeably slow disease progression and can cause severe complications, limiting their effectiveness. In addition to therapies targeting the core pathology of AD, a more comprehensive approach may be needed for its treatment. In recent years, non-pharmacological treatments such as physical therapy, exercise therapy, cell therapy, and nanoparticles have shown great potential in mitigating disease progression and alleviating clinical symptoms. This article reviews recent advances in non-pharmacological treatment approaches for AD, highlighting their contributions to AD management and facilitating the exploration of novel therapeutic strategies.
Collapse
Affiliation(s)
- Shaofen Wang
- West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
| | - Haochen Xu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guangdong Liu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Limei Chen
- West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
| |
Collapse
|
22
|
Lee K, Kim M. Evolutionary Insights into Irisin/FNDC5: Roles in Aging and Disease from Drosophila to Mammals. Biomolecules 2025; 15:261. [PMID: 40001564 PMCID: PMC11853655 DOI: 10.3390/biom15020261] [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] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The Irisin/FNDC5 protein family has emerged as a pivotal link between exercise and the prevention of age-associated diseases. Irisin is highly expressed during exercise from skeletal and cardiac muscle cells, playing a critical role in mediating systemic health benefits through its actions on various tissues. However, Irisin levels decline with age, correlating with a heightened incidence of diseases such as muscle weakness, cardiovascular disorders, and neurodegeneration. Notably, the administration of Irisin has shown significant potential in both preventing and treating these conditions. Recently, an Irisin/FNDC5 homolog was identified in an invertebrate Drosophila model, providing valuable insights into its conserved role in exercise physiology. Importantly, Irisin/FNDC5 has been demonstrated to regulate autophagy-a process essential for clearing excessive nutrients, toxic aggregates, and dysfunctional organelles-in both flies and mammals. Dysregulated autophagy is often implicated in age-related diseases, highlighting its relevance to Irisin/FNDC5's functions. These findings deepen our understanding of Irisin/FNDC5's roles and its potential as a therapeutic target for mitigating aging-related health decline. Further studies are needed to elucidate the precise mechanisms by which Irisin regulates autophagy and its broader impact on physiological aging and related diseases.
Collapse
Affiliation(s)
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
23
|
Zhang L, Peng Y, Kong Y, Zhang X, Li Z, Jia H. Circulating irisin levels in patients with sarcopenia: a systematic review and meta-analysis. Eur Geriatr Med 2025; 16:5-13. [PMID: 39562482 DOI: 10.1007/s41999-024-01097-5] [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: 03/14/2024] [Accepted: 10/24/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVE During the aging process, a decrease in irisin levels is associated with numerous bone and muscle diseases. This study aims to provide evidence of circulating irisin levels in patients with sarcopenia. METHODS This systematic review was conducted based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standard and the recommendations of the Cochrane Collaboration. A comprehensive search was conducted in PubMed, Embase, Web of Science databases, and other sources from their establishment until August 2023. The Review Manager software version 5.4 was used to calculate the standard mean difference (SMD). I2 statistics measured heterogeneity. RESULTS 12 studies involving 2133 participants who met the inclusion criteria were analyzed. We found that irisin levels were significantly lower in patients with sarcopenia (SMD: - 1.28; 95% CI - 1.65, - 0.90; I2 = 92% P < 0.001). Sensitivity analysis confirmed the robustness of this result. The correlation results showed that there was a positive correlation between the levels of circulating irisin and muscle mass (r value 0.62, 95% CI 0.31, 0.81; P < 0.001) and strength (r value 0.47, 95% CI 0.23, 0.66; P < 0.001), but no statistical correlation between irisin and muscle function (The P-values for gait speed and chair test time are 0.5523 and 0.1467, respectively). CONCLUSION No matter the study area, study design, blood samples, or diagnostic criteria, the concentration of circulating irisin in patients with sarcopenia was lower than that in the control group.
Collapse
Affiliation(s)
- Liangchuan Zhang
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yating Peng
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yuan Kong
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Xue Zhang
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Zetian Li
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Hong Jia
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China.
- Collaborating Center of the National Institute of Health Data Sciences of China, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
24
|
Cheon J, Kwon S, Kim M. Exerkines mitigating Alzheimer's disease progression by regulating inflammation: Focusing on macrophage/microglial NLRP3 inflammasome pathway. Alzheimers Dement 2025; 21:e14432. [PMID: 39641407 PMCID: PMC11848186 DOI: 10.1002/alz.14432] [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: 08/23/2024] [Revised: 10/16/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Recent research highlights the critical role of inflammation in accelerating amyloid beta and phosphorylated tubulin-associated protein tau cascade and Alzheimer's disease (AD) progression. Emerging evidence suggests that exercise influences AD by modulating inflammatory responses. We conducted a comprehensive search across multiple online databases. Our approach focused on previous and recent studies exploring the links among inflammation, AD, and the effects of exercise, specifically targeting research articles and books published in English. We pointed out that inflammation extends from the periphery to the central nervous system, facilitated by macrophage/microglial NLRP3 (nucleotide-binding domain, leucine rich-containing family, pyrin domain-containing protein 3) inflammasome signaling, which exacerbates classical AD mechanisms. Moreover, we provided further insights into the modulation of inflammasome signaling through exercise and exerkines, which may contribute to mitigating AD development. These insights deepen our understanding of AD mechanisms and offer the potential for identifying key therapeutic targets and biomarkers crucial for effective disease management and treatment. HIGHLIGHTS: Inflammation is potentially linked to the acceleration of classical Alzheimer's disease (AD) pathogenesis, including the pathways involving amyloid beta and phosphorylated tau, mediated by pro-inflammatory cytokines. Inflammation, initiated by the nucleotide-binding domain, leucine rich-containing family, pyrin domain-containing protein 3 (NLRP3) inflammasome signaling pathway within M1-type macrophages/microglia, may contribute to neuroinflammation and AD progression. Exercise has the potential to reduce inflammation and the development of AD by influencing NLRP3 inflammasome signaling via exerkines.
Collapse
Affiliation(s)
- Jaehwan Cheon
- Department of Biomedical ScienceKorea University College of MedicineSeongbuk‐guSeoulRepublic of Korea
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
| | - Soonyong Kwon
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
- Department of Chemistry & Life ScienceSahmyook UniversityNowon‐guSeoulRepublic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
- Department of Chemistry & Life ScienceSahmyook UniversityNowon‐guSeoulRepublic of Korea
| |
Collapse
|
25
|
Bigliassi M, Cabral DF, Evans AC. Improving brain health via the central executive network. J Physiol 2025. [PMID: 39856810 DOI: 10.1113/jp287099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Cognitive and physical stress have significant effects on brain health, particularly through their influence on the central executive network (CEN). The CEN, which includes regions such as the dorsolateral prefrontal cortex, anterior cingulate cortex and inferior parietal lobe, is central to managing the demands of cognitively challenging motor tasks. Acute stress can temporarily reduce connectivity within the CEN, leading to impaired cognitive function and emotional states. However a rebound in these states often follows, driven by motivational signals through the mesocortical and mesolimbic pathways, which help sustain inhibitory control and task execution. Chronic exposure to physical and cognitive challenges leads to long-term improvements in CEN functionality. These changes are supported by neurochemical, structural and systemic adaptations, including mechanisms of tissue crosstalk. Myokines, adipokines, anti-inflammatory cytokines and gut-derived metabolites contribute to a biochemical environment that enhances neuroplasticity, reduces neuroinflammation and supports neurotransmitters such as serotonin and dopamine. These processes strengthen CEN connectivity, improve self-regulation and enable individuals to adopt and sustain health-optimizing behaviours. Long-term physical activity not only enhances inhibitory control but also reduces the risk of age-related cognitive decline and neurodegenerative diseases. This review highlights the role of progressive physical stress through exercise as a practical approach to strengthening the CEN and promoting brain health, offering a strategy to improve cognitive resilience and emotional well-being across the lifespan.
Collapse
Affiliation(s)
- Marcelo Bigliassi
- Department of Teaching and Learning, Florida International University, Miami, Florida, USA
| | - Danylo F Cabral
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Amanda C Evans
- Functional Flow Solutions LLC, Albuquerque, New Mexico, USA
| |
Collapse
|
26
|
Tang N, Li W, Shang H, Yang Z, Chen Z, Shi G. Irisin-mediated KEAP1 degradation alleviates oxidative stress and ameliorates pancreatitis. Immunol Res 2025; 73:37. [PMID: 39821708 DOI: 10.1007/s12026-024-09588-0] [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: 10/16/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Oxidative stress (OS) injury is pivotal in acute pancreatitis (AP) pathogenesis, contributing to inflammatory cascades. Irisin, a ubiquitous cytokine, exhibits antioxidant properties. However, the role of irisin in AP remains inconclusive. Our study aims to elucidate irisin expression in AP patients and investigate its mechanism of action to propose a novel treatment strategy for AP. Serum irisin levels in 65 AP patients were quantified using an enzyme-linked immunosorbent assay and correlated with disease severity scores. Core genes implicated in AP-related oxidative stress were identified and screened via bioinformatics analysis. The therapeutic efficacy of irisin in AP was confirmed using a murine cerulein-induced AP model. The intrinsic mechanism of irisin's antioxidative stress action was investigated and verified in pancreatic AR42J cells (Supplementary Fig. 1). Common targets shared by irisin and AP were further validated using a molecular docking model which was constructed for virtual docking analysis. This study investigated alterations in redox status in AP and found a significant reduction in serum irisin levels, correlating inversely with AP severity. In a murine AP model, we showed that irisin triggers an antioxidative stress program via the KEAP1 gene; this process helps reestablish redox balance by decreasing the buildup of reactive oxygen species (ROS) and suppressing the secretion of inflammatory mediators within pancreatic tissues Notably, increased KEAP1 expression counteracted the antioxidative effects of irisin. Our findings unveil a novel therapeutic mechanism for AP, wherein irisin inhibits KEAP1 to alleviate OS. Increasing irisin levels in vivo presents a promising strategy for AP treatment.
Collapse
Affiliation(s)
- Nan Tang
- Dalian Medical University, Dalian, Liaoning, China
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Wendi Li
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Hezhen Shang
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Zhen Yang
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Zengyin Chen
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Guangjun Shi
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China.
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
27
|
López-Ojeda W, Hurley RA. Myokines and the Brain: A Novel Neuromuscular Endocrine Loop. J Neuropsychiatry Clin Neurosci 2025; 37:A4-4. [PMID: 39812655 DOI: 10.1176/appi.neuropsych.20240173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Affiliation(s)
- Wilfredo López-Ojeda
- Veterans Affairs Mid-Atlantic Mental Illness Research, Education and Clinical Center and the Research and Academic Affairs Service Line, W. G. Hefner Veterans Affairs Medical Center, Salisbury, N.C. (López-Ojeda, Hurley); Department of Psychiatry and Behavioral Medicine (López-Ojeda, Hurley) and Department of Radiology (Hurley), Wake Forest University School of Medicine, Winston-Salem, N.C
| | - Robin A Hurley
- Veterans Affairs Mid-Atlantic Mental Illness Research, Education and Clinical Center and the Research and Academic Affairs Service Line, W. G. Hefner Veterans Affairs Medical Center, Salisbury, N.C. (López-Ojeda, Hurley); Department of Psychiatry and Behavioral Medicine (López-Ojeda, Hurley) and Department of Radiology (Hurley), Wake Forest University School of Medicine, Winston-Salem, N.C
| |
Collapse
|
28
|
Campbell TS, Donoghue K, Roth TL. Gene Expression After Exercise Is Disrupted by Early-Life Stress. Dev Psychobiol 2025; 67:e70017. [PMID: 39780028 PMCID: PMC11711301 DOI: 10.1002/dev.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Exercise can be leveraged as an important tool to improve neural and psychological health, either on its own or to bolster the efficacy of evidence-based treatment modalities. Research in both humans and animal models shows that positive experiences, such as exercise, promote neuroprotection while, in contrast, aversive experiences, particularly those in early development, are often neurologically and psychologically disruptive. In the current study, we employed a preclinical model to investigate the therapeutic benefits of exercise on gene expression in the brains of adult rats. Long Evans rats were exposed to maltreatment stress or nurturing care during infancy, with some rats later given voluntary running wheels as an aerobic exercise intervention from Postnatal Days 70 to 90. Our results showed that irisin gene expression, which promotes neuroprotection, was differentially affected by exercise and early exposure to stress. We add to a rapidly growing area of research on the neuroprotective benefits of exercise and shed light on important molecular mechanisms that may affect the efficacy of exercise in different individuals.
Collapse
Affiliation(s)
- Taylor S. Campbell
- Department of Psychological & Brain SciencesUniversity of DelawareNewarkDelawareUSA
| | - Katelyn Donoghue
- Department of Psychological & Brain SciencesUniversity of DelawareNewarkDelawareUSA
| | - Tania L. Roth
- Department of Psychological & Brain SciencesUniversity of DelawareNewarkDelawareUSA
| |
Collapse
|
29
|
Shen C, Wu K, Ke Y, Zhang Q, Chen S, Li Q, Ruan Y, Yang X, Liu S, Hu J. Circulating irisin levels in patients with MAFLD: an updated systematic review and meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1464951. [PMID: 39741878 PMCID: PMC11686449 DOI: 10.3389/fendo.2024.1464951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/18/2024] [Indexed: 01/03/2025] Open
Abstract
Objective Current research suggests that irisin is closely linked to the pathogenesis and progression of metabolic dysfunction-associated fatty liver disease (MAFLD). This systematic review and meta-analysis updates our previous meta-analysis and further explores the relevance between circulating irisin levels and MAFLD. Methods Nine databases (PubMed, EMBASE, Cochrane Library, CNKI, Wanfang, Weipu, CBM, Clinicaltrials.gov and gray literature) were retrieved as of 1st August, 2024. The standardized mean difference (SMD) and 95% confidence interval (CI) represent pooled effect size. We used the Newcastle-Ottawa Scale to evaluate the quality of articles and the certainty of evidence assessed by GRADE system. All statistical analyses were performed using RevMan 5.3 and Stata 12(Stata Corporation, yi TX). Results Fifteen case-control studies were included. Circulating irisin levels in the MAFLD group were markedly lower than those in the healthy group (SMD=-1.04 [-1.93, -0.14]). Subgroup analyses by race, age, severity and T2DM revealed that circulating irisin levels were lower in the MAFLD group compared to those in the healthy controls in the Asian population (SMD=-1.38 [-2.44, -0.31], P<0.05) and in those above 50 years old (SMD=-2.23 [-3.64, -0.81], P<0.05) and higher in the mild MAFLD groups than those in moderate to severe MAFLD groups (SMD = 11.68 [9.05, 14.31], P<0.05). And the circulating irisin levels in MAFLD patients with T2DM were significantly lower than those in healthy group (SMD = -2.90 [-4.49, -1.30]). ELISA kits from different companies also presented different relationships. Conclusions There were significantly lower circulating irisin levels in the MAFLD group than in the healthy control group. Although these results differed from our previous results, there is no denying that circulating irisin levels are closely associated with the advancement of MAFLD.
Collapse
Affiliation(s)
- Chenglu Shen
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kaihan Wu
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yani Ke
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qin Zhang
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuaihang Chen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qicong Li
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuting Ruan
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xudan Yang
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shan Liu
- Department of Clinical Evaluation Center, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jie Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
30
|
Zhang QX, Zhang LJ, Zhao N, Yang L. Irisin in ischemic stroke, Alzheimer's disease and depression: a Narrative Review. Brain Res 2024; 1845:149192. [PMID: 39214327 DOI: 10.1016/j.brainres.2024.149192] [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: 06/02/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Irisin is a glycosylated protein formed from the hydrolysis of fibronectin type III domain-containing protein 5 (FNDC5). Irisin is widely involved in the regulation of glucose and lipid metabolism. In addition, recent studies have demonstrated that Irisin can inhibit inflammation, restrain oxidative stress and have neuroprotective effects, which suggests that Irisin may have a good therapeutic effect on central nervous system diseases. Therefore, this review summarizes the role of Irisin in central nervous system diseases, including its signal pathways and possible mechanisms, etc. Irisin may be a potential candidate drug for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Qiu-Xia Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Lin-Jie Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Ning Zhao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Li Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China.
| |
Collapse
|
31
|
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
|
32
|
Kostka M, Morys J, Małecki A, Nowacka-Chmielewska M. Muscle-brain crosstalk mediated by exercise-induced myokines - insights from experimental studies. Front Physiol 2024; 15:1488375. [PMID: 39687518 PMCID: PMC11647023 DOI: 10.3389/fphys.2024.1488375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Over the past couple of decades, it has become apparent that skeletal muscles might be engaged in endocrine signaling, mostly as a result of exercise or physical activity in general. The importance of this phenomenon is currently studied in terms of the impact that exercise- or physical activity -induced signaling factors have, in the interaction of the "muscle-brain crosstalk." So far, skeletal muscle-derived myokines were demonstrated to intercede in the connection between muscles and a plethora of various organs such as adipose tissue, liver, or pancreas. However, the exact mechanism of muscle-brain communication is yet to be determined. It is speculated that, in particular, brain-derived neurotrophic factor (BDNF), irisin, cathepsin B (CTSB), interleukin 6 (IL-6), and insulin-like growth factor-1 (IGF-1) partake in this crosstalk by promoting neuronal proliferation and synaptic plasticity, also resulting in improved cognition and ameliorated behavioral alterations. Researchers suggest that myokines might act directly on the brain parenchyma via crossing the blood-brain barrier (BBB). The following article reviews the information available regarding rodent studies on main myokines determined to cross the BBB, specifically addressing the association between exercise-induced myokine release and central nervous system (CNS) impairments. Although the hypothesis of skeletal muscles being critical sources of myokines seems promising, it should not be forgotten that the origin of these factors might vary, depending on the cell types engaged in their synthesis. Limited amount of research providing information on alterations in myokines expression in various organs at the same time, results in taking them only as circumstantial evidence on the way to determine the actual involvement of skeletal muscles in the overall state of homeostasis. The following article reviews the information available regarding rodent studies on main myokines determined to cross the BBB, specifically addressing the association between exercise-induced myokine release and CNS impairments.
Collapse
Affiliation(s)
| | | | | | - Marta Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
| |
Collapse
|
33
|
Villamil-Parra W, Moscoso-Loaiza L. Effects of physical exercise on Irisin and BDNF concentrations, and their relationship with cardiometabolic and mental health of individuals with Metabolic Syndrome: A Systematic Review. Exp Gerontol 2024; 198:112640. [PMID: 39579805 DOI: 10.1016/j.exger.2024.112640] [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: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Chronic Non-Communicable Diseases (NCDs), including cardiovascular diseases, cancer, chronic respiratory diseases, and diabetes, are the leading global causes of mortality, accounting for 71 % of deaths annually. Metabolic Syndrome (MS), characterized by hypertension, obesity, insulin resistance, and dyslipidemia, is a significant risk factor for NCDs. Physical inactivity exacerbates these conditions, contributing to poor cardiovascular and mental health outcomes. OBJECTIVE To analyze the effects of physical exercise on Irisin and Brain-Derived Neurotrophic Factor (BDNF) concentrations and their relationship with cardiometabolic and mental health of individuals with MS. METHODS A systematic review was conducted of articles published between August 2023 and June 2024 in ScienceDirect, PubMed, and SciELO, following PRISMA guidelines. Inclusion criteria encompassed observational studies, clinical trials, and reviews with high methodological quality. The review focused on Irisin, BDNF, physical exercise, and MS. RESULTS A total of 584 articles were identified, with 43 selected for detailed analysis. The review highlights that physical exercise significantly impacts Irisin and BDNF levels, which in turn influence metabolic and mental health. Irisin, a myokine secreted during exercise, promotes the conversion of white adipose tissue to brown adipose tissue, enhancing energy expenditure and metabolic health. Elevated Irisin levels are associated with improved cognitive function and mental well-being. BDNF, a neurotrophin, supports neuronal growth and cognitive function. Exercise-induced increases in BDNF levels are linked to enhanced neuroplasticity, reduced anxiety, and improved mood. CONCLUSION Understanding the role of Irisin and BDNF in response to physical exercise offers valuable insights for developing strategies to manage and prevent MS and its related mental health issues. Further research is needed to elucidate the molecular mechanisms involved.
Collapse
Affiliation(s)
- Wilder Villamil-Parra
- Departamento de Movimiento Corporal Humano, Facultad de Enfermería y Rehabilitación, Universidad de la Sabana, Puente del Común Km. 7, Autopista Norte, Chía, Cundinamarca, Colombia.
| | - Luisa Moscoso-Loaiza
- Departamento de Enfermería, Facultad de Enfermería, Universidad Nacional de Colombia, Sede Bogotá, Carrera 30 No. 45-03 Bogotá, D.C., Colombia.
| |
Collapse
|
34
|
Dai S, Dong Y, Shi H, Jin J, Gan Y, Li X, Wu Y, Wang F, Zhu X, Hu Q, Dong Y, Fu Y. Aerobic exercise prevents and improves cognitive dysfunction caused by morphine withdrawal via regulating endogenous opioid peptides in the brain. Psychopharmacology (Berl) 2024; 241:2525-2537. [PMID: 39417859 DOI: 10.1007/s00213-024-06698-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Morphine withdrawal leads to serious cognitive deficits in which dynorphins are directly involved. Recently, exercise has been shown to prevent and improve cognition dysfunction in a variety of ways. Meanwhile, exercise can regulate the endogenous opioid peptides including dynorphins. However, it remains unclear whether exercise influences cognitive dysfunction caused by morphine withdrawal via dynorphins. In the current study, we investigate the physiological mechanism of exercise prevention and improvement aganist cognition dysfunction caused by morphine withdrawal. METHODS Male, adult C57BL/6 mice were randomly divided into 5 groups : Saline control (WT), exercise (EXE), morphine withdrawl (MW), exercise + morphine withdrawl (EMW), morphine withdrawl + exercise (MWE). We established aerobic exercise prevention/improvement models, and conducted behavioral tests including Open field test (OFT), Temporal order memory test (TOM) and Y-maze. Through Western Blotting and immunofluorescence staining, we detected endogenous opioid peptides in hippocampus and mPFC. RESULTS Compared with MW group, EMW group and MWE group showed the same performance as WT group in TOM and Y-maze, with correct object recognition and memory ability. In Western Blotting and immunofluorescence staining experiments, it indicated that EMW group reduced the expression of PDYN and its fluorescence intensity in hippocampus; MWE group reduced the expression of OPRK1 and its fluorescence intensity in mPFC. CONCLUSION Our data suggest that aerobic exercise can both prevent and improve cognitive dysfunction caused by acute morphine withdrawal via respectively down-regulating PDYN in the hippocampus and down-regulating OPRK1 in the mPFC. They may become new targets for drugs development in the future.
Collapse
Affiliation(s)
- Shanghua Dai
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Yigang Dong
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Haifeng Shi
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Jiawei Jin
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Yixia Gan
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Xinyi Li
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Yongkang Wu
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Fanglin Wang
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Xinrui Zhu
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Qingmiao Hu
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Yi Dong
- Key Laboratory of Adolescent HealthAssessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, 200241, China.
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China.
| | - Yingmei Fu
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
35
|
Wang YT, Zheng SY, Jiang SD, Luo Y, Wu YX, Naranmandakh S, Li YS, Liu SG, Xiao WF. Irisin in degenerative musculoskeletal diseases: Functions in system and potential in therapy. Pharmacol Res 2024; 210:107480. [PMID: 39490914 DOI: 10.1016/j.phrs.2024.107480] [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: 08/16/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Degenerative musculoskeletal diseases are a class of diseases related to the gradual structural and functional deterioration of muscles, joints, and bones, including osteoarthritis (OA), osteoporosis (OP), sarcopenia (SP), and intervertebral disc degeneration (IDD). As the proportion of aging people around the world increases, degenerative musculoskeletal diseases not only have a multifaceted impact on patients, but also impose a huge burden on the medical industry in various countries. Therefore, it is crucial to find key regulatory factors and potential therapeutic targets. Recent studies have shown that irisin plays an important role in degenerative musculoskeletal diseases, suggesting that it may become a key molecule in the prevention and treatment of degenerative diseases of the musculoskeletal system. Therefore, this review provides a comprehensive description of the release and basic functions of irisin, and summarizes the role of irisin in OA, OP, SP, and IDD from a cellular and tissue perspective, providing comprehensive basis for clinical application. In addition, we summarized the many roles of irisin as a key information molecule in bone-muscle-adipose crosstalk and a regulatory molecule involved in inflammation, senescence, and cell death, and proposed the interesting possibility of irisin in degenerative musculoskeletal diseases.
Collapse
Affiliation(s)
- Yu-Tong Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Sheng-Yuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shi-de Jiang
- The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Yan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Yu-Xiang Wu
- School of Kinesiology, Jianghan University, Wuhan, Hubei, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shu-Guang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Wen-Feng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
36
|
Wang LY, Hu H, Sheng ZH, Hu HY, Zhang ZH, Tan L. Associations among healthy lifestyle characteristics, neuroinflammation, and cerebrospinal fluid core biomarkers of Alzheimer's disease in cognitively intact adults: The CABLE study. J Alzheimers Dis 2024; 102:855-865. [PMID: 39558781 DOI: 10.1177/13872877241291969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
BACKGROUND The occurrence of Alzheimer's disease (AD) can be partially prevented through healthy lifestyles, but the mechanisms associated with AD pathology are unclear. OBJECTIVE To explore associations among healthy lifestyle characteristics (HLCs), cerebrospinal fluid (CSF) soluble TREM2 (sTREM2), and AD biomarkers. METHODS From the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study, 924 cognitively normal participants were enrolled in this cross-sectional analysis. We defined the following 11 HLCs: appropriate frequencies of coffee and tea consumption, sufficient frequencies of fish and fruit intake, non-social isolation, adequate sleep, regular physical activity, no depression, never smoking, non-hazardous drinking, and well-maintained blood pressure. We categorized participants according to the number of HLCs reported by participants into favorable, intermediate, and unfavorable lifestyle groups. Multiple linear regression was used to investigate the relationship among HLCs, CSF sTREM2, and AD biomarkers. Mediation effects were tested using a causal mediation analysis having 10,000 bootstrap iterations. RESULTS Included subjects were with a mean age of 61.8 ± 10.2 years, of which 41.8% were female. Sufficient fish intake (β = -0.164, p = 0.017) and well-maintained blood pressure (β = -0.232, p = 0.006) were significantly correlated with lower CSF sTREM2 levels. A larger number of HLCs were associated with lower CSF T-tau (p = 0.001), P-tau (p = 0.012), and sTREM2 (p = 0.040) levels. CSF sTREM2 partially mediated the association between the number of HLCs and CSF tau pathology (mediating proportion T-tau: 22.4%; P-tau: 25.0%). CONCLUSIONS HLCs might impact the pathological processes of AD by regulating neuroinflammation.
Collapse
Affiliation(s)
- Lan-Yang Wang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ze-Hu Sheng
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zi-Hao Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
De Sousa RAL. Exercise-produced irisin effects on brain-related pathological conditions. Metab Brain Dis 2024; 39:1679-1687. [PMID: 39145861 DOI: 10.1007/s11011-024-01412-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Exercise increases peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α) expression, which in turn causes the fibronectin type III domain containing 5 (FNDC5) protein to be produced. This protein is then cleaved, primarily in skeletal muscle fibers, to produce irisin. When the mature FNDC5 is cleaved by proteases, Irisin - which is the fibronectin III domain without the signal sequence - is released. Resistance, aerobic, and high-intensity interval training (HIIT) are recognized as forms of physical exercise that raise irisin levels, and insulin receptor phosphorylation in tyrosine residues, favoring an increase in the activity of the insulin-dependent pathway (PI3K pathway) and assisting in the fight against insulin resistance, inflammation, and cognitive decline. Irisin may represent a promising option for the therapeutic targeting in several brain-related pathological conditions, like Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, type 2 diabetes, and obesity. Exercise protocols are healthy and inexpensive interventions that can help find cellular and molecular changes in several brain-related pathological conditions. Here, it was reviewed what is known about exercise-produced irisin studies involving AD, PD, epilepsy, type 2 diabetes, and obesity.
Collapse
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Physical Education Department, Federal University of the Valleys of Jequitinhonha and Mucuri (UFVJM), Diamantina, MG, Brazil.
- Neuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício - GENE), UFVJM, Diamantina, MG, Brazil.
- Multicenter Graduate Program in Physiological Sciences, Laboratório Experimental de Treinamento Físico (LETFIS), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Rodovia MGT 367, Km 583, Alto da Jacuba, nº 5000, Diamantina, MG, CEP 39100-000, Brazil.
| |
Collapse
|
38
|
Lu X, Xiong W, Chen Z, Li Y, Xu F, Yang X, Long M, Guo W, Wu S, Sun L, Wang G. Exercise-conditioned plasma ameliorates postoperative cognitive dysfunction by activating hippocampal cholinergic circuit and enhancing BDNF/TrkB signaling. Cell Commun Signal 2024; 22:551. [PMID: 39558340 PMCID: PMC11572510 DOI: 10.1186/s12964-024-01938-7] [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: 06/18/2024] [Accepted: 11/10/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a prevalent complication following anesthesia and surgery, particularly in the elderly, leading to increased mortality and reduced quality of life. Despite its prevalence, there are no effective clinical treatments. Exercise has shown cognitive benefits in aging and various diseases, which can be transferred to sedentary animals through plasma. However, it is unclear if exercise-conditioned plasma can replicate these benefits in the context of POCD. METHODS Sixteen-month-old male C57BL/6J mice underwent 30 days of voluntary running wheel training or received systemic administration of exercise-conditioned plasma, followed by tibial fracture surgery under general anesthesia at 17 months of age. Cognitive performance, hippocampal synaptic deficits, neuroinflammation, BDNF/TrkB signaling, and medial septum (MS)-hippocampal cholinergic activity were evaluated through immunohistochemical staining, transmission electron microscopy, Western blotting, and biochemical assays. To investigate the role of hippocampal BDNF signaling and cholinergic activity in the therapeutic effects, the TrkB antagonist ANA-12 and the cholinergic receptor muscarinic 1 (CHRM1) antagonist trihexyphenidyl (THP) were administered via intraperitoneal injection, and adeno-associated virus (AAV) vectors expressing Chrm1 shRNA were delivered via intrahippocampal stereotaxic microinjection. RESULTS Exercise-conditioned plasma mimicked the benefits of exercise, alleviating cognitive decline induced by anesthesia/surgery, restoring hippocampal synapse formation and levels of regulators for synaptic plasticity, inhibiting neuroinflammatory responses to surgery by microglia and astrocytes, augmenting BDNF production and TrkB phosphorylation in hippocampal neurons, astrocytes, and microglia, upregulating MS expression of choline acetyltransferase (CHAT) and hippocampal expression of CHRM1 in neurons and astrocytes, and enhancing hippocampal cholinergic innervation and acetylcholine release. Conversely, ANA-12 administration blocked TrkB activation and reduced the protective effects on cognition, synaptic deficits, and neuroinflammatory reactivity of glial cells post-surgery. Similarly, THP administration or intrahippocampal delivery of AAV-Chrm1 shRNA inhibited the activation of the hippocampal cholinergic circuit by exercise plasma, negating the cognitive and neuropathological benefits and reducing BDNF/TrkB signaling enhancements. CONCLUSION Exercise-conditioned plasma can replicate the protective effects of exercise against anesthesia/surgery-induced neuroinflammation, synaptic, and cognitive impairments, at least partly, through CHRM1-dependent regulation of hippocampal cholinergic activity and BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Xiaodi Lu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Weijie Xiong
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Zhuo Chen
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yurou Li
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Fengyan Xu
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Xue Yang
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Meiwen Long
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Wenhan Guo
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Shuliang Wu
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China.
| | - Liang Sun
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China.
| | - Guonian Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
39
|
Qiu R, Sun W, Su Y, Sun Z, Fan K, Liang Y, Lin X, Zhang Y. Irisin's emerging role in Parkinson's disease research: A review from molecular mechanisms to therapeutic prospects. Life Sci 2024; 357:123088. [PMID: 39357796 DOI: 10.1016/j.lfs.2024.123088] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder characterized by impaired motor function, is typically treated with medications and surgery. However, recent studies have validated physical exercise as an effective adjunct therapy, significantly improving both motor and non-motor symptoms in PD patients. Irisin, a myokine, has garnered increasing attention for its beneficial effects on the nervous system. Research has shown that irisin plays a crucial role in regulating metabolic balance, optimizing autophagy, maintaining mitochondrial quality, alleviating oxidative stress and neuroinflammation, and regulating cell death-all processes intricately linked to the pathogenesis of PD. This review examines the mechanisms through which irisin may counteract PD, provides insights into its biological effects, and considers its potential as a target for therapeutic strategies.
Collapse
Affiliation(s)
- Ruqing Qiu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Weilu Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yana Su
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Kangli Fan
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yue Liang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyue Lin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
40
|
Yang L, Zhou X, Heng T, Zhu Y, Gong L, Liu N, Yao X, Luo Y. FNDC5/Irisin mitigates high glucose-induced neurotoxicity in HT22 cell via ferroptosis. Biosci Trends 2024; 18:465-475. [PMID: 39414463 DOI: 10.5582/bst.2024.01249] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Diabetes-induced neuropathy represents a major etiology of dementia, highlighting an urgent need for the development of effective therapeutic interventions. In this study, we explored the role of fibronectin type III domain containing 5 (FNDC5)/Irisin in mitigating hyperglycemia-induced neurotoxicity in HT22 cells and investigated the underlying mechanisms. Our findings reveal that high glucose conditions are neurotoxic, leading to reduced viability of HT22 cells and increased apoptosis. Furthermore, the elevated expression of the intracellular ferroptosis marker Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4), along with increased levels of ferrous ions and malondialdehyde (MDA), suggests that high glucose conditions may induce ferroptosis in HT22 cells. FNDC5/Irisin treatment effectively mitigates high glucose-induced neurotoxicity and ferroptosis in HT22 cells. Mechanistically, FNDC5/Irisin enhances cellular antioxidant capacity, regulates ACSL4 expression, and improves intracellular redox status, thereby inhibiting ferroptosis and increasing HT22 cell survival under high-glucose conditions. These results highlight the neuroprotective potential of FNDC5/Irisin in high glucose environments, offering a promising avenue for developing treatments for diabetes-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Lingling Yang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaohan Zhou
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tian Heng
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yinghai Zhu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Rehabilitation, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, China
| | - Lihuan Gong
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Na Liu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiuqing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipality Clinical Research Center for Geriatric Medicine, Chongqing, China
- Department of Rehabilitation Therapy, Chongqing Medical University, Chongqing, China
| | - Yaxi Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
41
|
Nunkoo VS, Cristian A, Jurcau A, Diaconu RG, Jurcau MC. The Quest for Eternal Youth: Hallmarks of Aging and Rejuvenating Therapeutic Strategies. Biomedicines 2024; 12:2540. [PMID: 39595108 PMCID: PMC11591597 DOI: 10.3390/biomedicines12112540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The impressive achievements made in the last century in extending the lifespan have led to a significant growth rate of elderly individuals in populations across the world and an exponential increase in the incidence of age-related conditions such as cardiovascular diseases, diabetes mellitus type 2, and neurodegenerative diseases. To date, geroscientists have identified 12 hallmarks of aging (genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, impaired macroautophagy, mitochondrial dysfunction, impaired nutrient sensing, cellular senescence, stem cell exhaustion, defective intercellular communication, chronic inflammation, and gut dysbiosis), intricately linked among each other, which can be targeted with senolytic or senomorphic drugs, as well as with more aggressive approaches such as cell-based therapies. To date, side effects seriously limit the use of these drugs. However, since rejuvenation is a dream of mankind, future research is expected to improve the tolerability of the available drugs and highlight novel strategies. In the meantime, the medical community, healthcare providers, and society should decide when to start these treatments and how to tailor them individually.
Collapse
Affiliation(s)
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
42
|
Neira G, Hernández-Pardos AW, Becerril S, Ramírez B, Valentí V, Moncada R, Catalán V, Gómez-Ambrosi J, Burrell MA, Silva C, Escalada J, Frühbeck G, Rodríguez A. Differential mitochondrial adaptation and FNDC5 production in brown and white adipose tissue in response to cold and obesity. Obesity (Silver Spring) 2024; 32:2120-2134. [PMID: 39327772 DOI: 10.1002/oby.24132] [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: 04/24/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVE Fibronectin type III domain-containing protein 5 (FNDC5) modulates adipocyte metabolism by increasing white and brown adipose tissue (WAT and BAT) browning and activity, respectively. We investigated whether FNDC5 can regulate visceral WAT and BAT adaptive thermogenesis by improving mitochondrial homeostasis in response to cold and obesity. METHODS Adipose tissue expression of FNDC5 and factors involved in mitochondrial homeostasis were determined in patients with normal weight and obesity (n = 159) and in rats with diet-induced obesity after 1 week of cold exposure (n = 61). The effect of different FNDC5 concentrations on mitochondrial biogenesis, dynamics, and mitophagy was evaluated in vitro in human adipocytes. RESULTS In human visceral adipocytes, FNDC5/irisin triggered mitochondrial biogenesis (TFAM) and fusion (MFN1, MFN2, and OPA1) while inhibiting peripheral fission (DNM1L and FIS1) and mitophagy (PINK1 and PRKN). Circulating and visceral WAT expression of FNDC5 was decreased in patients and experimental animals with obesity, whereas its receptor, integrin αV, was upregulated. Obesity increased mitochondrial fusion while decreasing mitophagy in visceral WAT from patients and rats. By contrast, in rat BAT, an upregulation of Fndc5 and genes involved in mitochondrial biogenesis and fission was observed. Cold exposure promoted mitochondrial biogenesis and healthy peripheral fission while repressing Fndc5 expression and mitophagy in BAT from rats. CONCLUSIONS Depot differences in FNDC5 production and mitochondrial adaptations in response to obesity and cold might indicate a self-regulatory mechanism to control thermogenesis in response to energy needs.
Collapse
Affiliation(s)
- Gabriela Neira
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Víctor Valentí
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - María A Burrell
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Camilo Silva
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Escalada
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
43
|
Zhang H, Liang J, Huang J, Wang M, Wu L, Wu T, Chen N. Exerkine irisin mitigates cognitive impairment by suppressing gut-brain axis-mediated inflammation. J Adv Res 2024:S2090-1232(24)00485-5. [PMID: 39481644 DOI: 10.1016/j.jare.2024.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/16/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024] Open
Abstract
INTRODUCTION Exercise has been recognized to improve cognitive performance by optimizing gut flora and up-regulating exerkine irisin. OBJECTIVE Although exercise-induced irisin is beneficial to cognitive improvement, whether this benefit is achieved by optimizing gut microbiota and metabolites is not fully explored. METHODS After aerobic exercise and exogenous irisin interventions for 12 weeks, the 16S rRNA and metabolites in feces of 21-month-old mice were analyzed. Meanwhile, the differential miRNAs and mRNAs in hippocampal tissues were screened by high-throughput sequencing. Relevant mRNAs and proteins were evaluated by RT-PCR, Western blot, and immunofluorescence. RESULTS Compared with the young control mice, irisin levels and cognitive capacity of aged mice revealed a significant reduction, while aerobic exercise and intraperitoneal injection of exogenous irisin reversed aging-induced cognitive impairment. Similarly, 147 up-regulated and 173 down-regulated metabolites were detected in aged mice, while 64 and 45 up-regulated and 225 and 187 down-regulated metabolites were detected in aged mice with exercise and irisin interventions, respectively. Moreover, during hippocampal miRNA and mRNA sequencing analysis, 9 differential gut flora and 35 differential genes were identified to be correlated with the inflammatory signaling mediated by the TLR4/MyD88 signal pathway. CONCLUSION Aging-induced cognitive impairment is due to insulin resistance induced by TLR4/MyD88 signaling activation in hippocampal tissues mediated by gut microbiota and metabolite changes. Myokine irisin may be an important mediator in optimizing gut microbiota and metabolism for an improved understanding of mitigated aging process upon exercise interventions.
Collapse
Affiliation(s)
- Hu Zhang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jiling Liang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jielun Huang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Minghui Wang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Liangwen Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Tong Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
44
|
Liu Y, Fu X, Zhao X, Cui R, Yang W. The role of exercise-related FNDC5/irisin in depression. Front Pharmacol 2024; 15:1461995. [PMID: 39484160 PMCID: PMC11524886 DOI: 10.3389/fphar.2024.1461995] [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: 07/09/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024] Open
Abstract
The complexity of depression presents a significant challenge to traditional treatment methods, such as medication and psychotherapy. Recent studies have shown that exercise can effectively reduce depressive symptoms, offering a new alternative for treating depression. However, some depressed patients are unable to engage in regular physical activity due to age, physical limitations, and other factors. Therefore, pharmacological agents that mimic the effects of exercise become a potential treatment option. A newly discovered myokine, irisin, which is produced during exercise via cleavage of its precursor protein fibronectin type III domain-containing protein 5 (FNDC5), plays a key role in regulating energy metabolism, promoting adipose tissue browning, and improving insulin resistance. Importantly, FNDC5 can promote neural stem cell differentiation, enhance neuroplasticity, and improve mood and cognitive function. This review systematically reviews the mechanisms of action of exercise in the treatment of depression, outlines the physiology of exercise-related irisin, explores possible mechanisms of irisin's antidepressant effects. The aim of this review is to encourage future research and clinical applications of irisin in the prevention and treatment of depression.
Collapse
Affiliation(s)
- Yaqi Liu
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xiying Fu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin, China
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xing Zhao
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Yang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
45
|
Xu H, Tian X, Wang Y, Lin J, Zhu B, Zhao C, Wang B, Zhang X, Sun Y, Li N, Sun X, Zeng F, Li M, Ya X, Zhao R. Exercise Promotes Hippocampal Neurogenesis in T2DM Mice via Irisin/TLR4/MyD88/NF-κB-Mediated Neuroinflammation Pathway. BIOLOGY 2024; 13:809. [PMID: 39452118 PMCID: PMC11504848 DOI: 10.3390/biology13100809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Neuroinflammation is a major feature of type 2 diabetic mellitus (T2DM), adversely affecting hippocampal neurogenesis. However, the precise mechanism is not fully understood, and therapeutic approaches are currently lacking. Therefore, we determined the effects of exercise on neuroinflammation and hippocampal neurogenesis in T2DM mice, with a specific focus on understanding the role of the irisin and related cascade pathways in modulating the beneficial effects of exercise in these processes. Ten-week exercise significantly decreased T2DM-induced inflammation levels and markedly promoted hippocampal neurogenesis and memory function. However, these positive effects were reversed by 10 weeks of treatment with cyclo RGDyk, an inhibitor of irisin receptor signaling. Additionally, exercise helped reduce the M1 phenotype polarization of hippocampal microglia in diabetic mice; this effect could be reversed with cyclo RGDyk treatment. Moreover, exercise markedly increased the levels of fibronectin type III domain-containing protein 5 (FNDC5)/irisin protein while decreasing the expression of Toll-like receptor 4 (TLR4), myeloid differential protein-88 (MyD88), and nuclear factor kappa-B (NF-κB) in the hippocampus of T2DM mice. However, blocking irisin receptor signaling counteracted the down-regulation of TLR4/MyD88/NF-κB in diabetic mice undergoing exercise intervention. Conclusively, exercise appears to be effective in reducing neuroinflammation and enhancing hippocampal neurogenesis and memory in diabetes mice. The positive effects are involved in the participation of the irisin/TLR4/MyD88/NF-κB signaling pathway, highlighting the potential of exercise in the management of diabetic-induced cognitive decline.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Renqing Zhao
- College of Physical Education, Yangzhou University, Yangzhou 225127, China; (H.X.); (X.T.); (Y.W.); (J.L.); (B.Z.); (C.Z.); (B.W.); (X.Z.); (Y.S.); (N.L.); (X.S.); (F.Z.); (M.L.); (X.Y.)
| |
Collapse
|
46
|
Jaqua EE, Tran MLN, Alvarez P, Gupta M, Yoong J. Dementia and Cognitive Decline: A HEALM Approach. Am J Lifestyle Med 2024:15598276241291508. [PMID: 39540186 PMCID: PMC11556629 DOI: 10.1177/15598276241291508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Dementia and cognitive decline pose significant global public health challenges, with prevalence expected to rise in the coming decades. Lifestyle medicine offers a promising approach to mitigating cognitive issues through six key interventions: diet, physical activity, restorative sleep, social connections, stress management, and avoiding risky substances. Traditional methods like randomized controlled trials (RCTs) have limitations in capturing the long-term impacts of these interventions. To overcome these challenges, the American College of Lifestyle Medicine (ACLM) and the True Health Initiative (THI) developed the Hierarchies of Evidence Applied to Lifestyle Medicine (HEALM) framework, informed by the Evidence Threshold Pathway Mapping (ETPM) approach. This framework integrates diverse evidence sources to assess intervention effects over time. Applying HEALM, this review evaluates lifestyle factors' impact on dementia and cognitive decline. It finds strong evidence supporting plant-based nutrition, physical activity, restorative sleep, and avoiding risky substances in promoting cognitive health. Social connections may mitigate cognitive decline, while stress management requires further investigation due to inconclusive findings. Integrating these findings into public health strategies could effectively address the growing dementia burden and enhance overall well-being in aging populations, underscoring the need for continued research in cognitive health.
Collapse
Affiliation(s)
- Ecler E. Jaqua
- Family Medicine Department, Loma Linda University School of Medicine, Loma Linda, CA, USA (EJ, MLT, PA, MG)
| | - Mai-Linh N. Tran
- Family Medicine Department, Loma Linda University School of Medicine, Loma Linda, CA, USA (EJ, MLT, PA, MG)
| | - Pedro Alvarez
- Family Medicine Department, Loma Linda University School of Medicine, Loma Linda, CA, USA (EJ, MLT, PA, MG)
| | - Monica Gupta
- Family Medicine Department, Loma Linda University School of Medicine, Loma Linda, CA, USA (EJ, MLT, PA, MG)
| | - Jessica Yoong
- Family Medicine Department, Kaiser Permanente Riverside Medical Center, Riverside, CA, USA (JY)
| |
Collapse
|
47
|
Li D, Jia J, Zeng H, Zhong X, Chen H, Yi C. Efficacy of exercise rehabilitation for managing patients with Alzheimer's disease. Neural Regen Res 2024; 19:2175-2188. [PMID: 38488551 PMCID: PMC11034587 DOI: 10.4103/1673-5374.391308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/16/2023] [Accepted: 11/25/2023] [Indexed: 04/24/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and degenerative neurological disease characterized by the deterioration of cognitive functions. While a definitive cure and optimal medication to impede disease progression are currently unavailable, a plethora of studies have highlighted the potential advantages of exercise rehabilitation for managing this condition. Those studies show that exercise rehabilitation can enhance cognitive function and improve the quality of life for individuals affected by AD. Therefore, exercise rehabilitation has been regarded as one of the most important strategies for managing patients with AD. Herein, we provide a comprehensive analysis of the currently available findings on exercise rehabilitation in patients with AD, with a focus on the exercise types which have shown efficacy when implemented alone or combined with other treatment methods, as well as the potential mechanisms underlying these positive effects. Specifically, we explain how exercise may improve the brain microenvironment and neuronal plasticity. In conclusion, exercise is a cost-effective intervention to enhance cognitive performance and improve quality of life in patients with mild to moderate cognitive dysfunction. Therefore, it can potentially become both a physical activity and a tailored intervention. This review may aid the development of more effective and individualized treatment strategies to address the challenges imposed by this debilitating disease, especially in low- and middle-income countries.
Collapse
Affiliation(s)
- Dan Li
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Jinning Jia
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Haibo Zeng
- Department of Pathology, Huichang County People’s Hospital, Ganzhou, Jiangxi Province, China
| | - Xiaoyan Zhong
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Chenju Yi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong Province, China
| |
Collapse
|
48
|
King C, Rogers LG, Jansen J, Sivayokan B, Neyhard J, Warnes E, Hall SE, Plakke B. Adolescent treadmill exercise enhances hippocampal brain-derived neurotrophic factor (BDNF) expression and improves cognition in autism-modeled rats. Physiol Behav 2024; 284:114638. [PMID: 39004196 PMCID: PMC12032843 DOI: 10.1016/j.physbeh.2024.114638] [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: 06/03/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by repetitive behaviors and altered communication abilities. Exercise is a low-cost intervention that could improve cognitive function and improve brain plasticity mechanisms. Here, the valproic acid (VPA) model was utilized to induce ASD-like phenotypes in rodents. Animals were exercised on a treadmill and performance was evaluated on a cognitive flexibility task. Biomarkers related to exercise and plasticity regulation were quantified from the prefrontal cortex, hippocampus, and skeletal muscle. Exercised VPA animals had higher levels of hippocampal BDNF compared to sedentary VPA animals and upregulated antioxidant enzyme expression in skeletal muscle. Cognitive improvements were demonstrated in both sexes, but in different domains of cognitive flexibility. This research demonstrates the benefits of exercise and provides evidence that molecular responses to exercise occur in both the central nervous system and in the periphery. These results suggest that improving regulation of BDNF via exercise, even at low intensity, could provide better synaptic regulation and cognitive benefits for individuals with ASD.
Collapse
Affiliation(s)
- Cole King
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Liza G Rogers
- Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Jeremy Jansen
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Bhavana Sivayokan
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Jenna Neyhard
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Ellie Warnes
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Stephanie E Hall
- Anatomy and Physiology, Kansas State University, Manhattan, KS, 66506, USA
| | - Bethany Plakke
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
49
|
Li R, Miao Z, Liu Y, Chen X, Wang H, Su J, Chen J. The Brain-Gut-Bone Axis in Neurodegenerative Diseases: Insights, Challenges, and Future Prospects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307971. [PMID: 39120490 PMCID: PMC11481201 DOI: 10.1002/advs.202307971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 06/04/2024] [Indexed: 08/10/2024]
Abstract
Neurodegenerative diseases are global health challenges characterized by the progressive degeneration of nerve cells, leading to cognitive and motor impairments. The brain-gut-bone axis, a complex network that modulates multiple physiological systems, has gained increasing attention owing to its profound effects on the occurrence and development of neurodegenerative diseases. No comprehensive review has been conducted to clarify the triangular relationship involving the brain-gut-bone axis and its potential for innovative therapies for neurodegenerative disorders. In light of this, a new perspective is aimed to propose on the interplay between the brain, gut, and bone systems, highlighting the potential of their dynamic communication in neurodegenerative diseases, as they modulate multiple physiological systems, including the nervous, immune, endocrine, and metabolic systems. Therapeutic strategies for maintaining the balance of the axis, including brain health regulation, intestinal microbiota regulation, and improving skeletal health, are also explored. The intricate physiological interactions within the brain-gut-bone axis pose a challenge in the development of effective treatments that can comprehensively target this system. Furthermore, the safety of these treatments requires further evaluation. This review offers a novel insights and strategies for the prevention and treatment of neurodegenerative diseases, which have important implications for clinical practice and patient well-being.
Collapse
Affiliation(s)
- Rong Li
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Zong Miao
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Yu'e Liu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Xiao Chen
- Department of OrthopedicsXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
- Institute of Translational MedicineShanghai UniversityShanghai200444China
- Organoid Research CenterShanghai UniversityShanghai200444China
| | - Hongxiang Wang
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Jiacan Su
- Department of OrthopedicsXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
- Institute of Translational MedicineShanghai UniversityShanghai200444China
- Organoid Research CenterShanghai UniversityShanghai200444China
| | - Juxiang Chen
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| |
Collapse
|
50
|
Li Q, Li C, Zhang X. Research Progress on the Effects of Different Exercise Modes on the Secretion of Exerkines After Spinal Cord Injury. Cell Mol Neurobiol 2024; 44:62. [PMID: 39352588 PMCID: PMC11445308 DOI: 10.1007/s10571-024-01497-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: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Exercise training is a conventional treatment strategy throughout the entire treatment process for patients with spinal cord injury (SCI). Currently, exercise modalities for SCI patients primarily include aerobic exercise, endurance training, strength training, high-intensity interval training, and mind-body exercises. These exercises play a positive role in enhancing skeletal muscle function, inducing neuroprotection and regeneration, thereby influencing neural plasticity, reducing limb spasticity, and improving motor function and daily living abilities in SCI patients. However, the mechanism by which exercise training promotes functional recovery after SCI is still unclear, and there is no consensus on a unified and standardized exercise treatment plan. Different exercise methods may bring different benefits. After SCI, patients' physical activity levels decrease significantly due to factors such as motor dysfunction, which may be a key factor affecting changes in exerkines. The changes in exerkines of SCI patients caused by exercise training are an important and highly relevant and visual evaluation index, which may provide a new research direction for revealing the intrinsic mechanism by which exercise promotes functional recovery after SCI. Therefore, this article summarizes the changes in the expression of common exerkines (neurotrophic factors, inflammatory factors, myokines, bioactive peptides) after SCI, and intends to analyze the impact and role of different exercise methods on functional recovery after SCI from the perspective of exerkines mechanism. We hope to provide theoretical basis and data support for scientific exercise treatment programs after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|