Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Aging Neurosci., 26 August 2025

Sec. Parkinson’s Disease and Aging-related Movement Disorders

Volume 17 - 2025 | https://doi.org/10.3389/fnagi.2025.1580029

Exercise as a multitarget therapy: modulating myokines, neurotrophins, and inflammation in Parkinson’s disease


Wei-Qi Li,Wei-Qi Li1,2Jia-Hua Yang,Jia-Hua Yang1,2Lu-Lu Liu,Lu-Lu Liu1,2Song-Tao DingSong-Tao Ding1Bin YuBin Yu1Lin JiangLin Jiang1Ning YanNing Yan3Han-Deng Liu,,*Han-Deng Liu1,4,5*
  • 1Laboratory of Tissue and Cell Biology, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, China
  • 2College of First Clinical, Chongqing Medical University, Chongqing, China
  • 3Department of Neurology, University-Town Hospital of Chongqing Medical University, Chongqing, China
  • 4Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
  • 5Center for Neuroscience Research, Chongqing Medical University, Chongqing, China

Parkinson’s disease is a progressive neurodegenerative disorder characterized by degeneration of dopaminergic neurons, leading to significant motor and non-motor symptoms. Recent studies emphasize that exercise is a beneficial intervention, not only helping to decrease the risk of developing of Parkinson’s disease but also alleviating existing symptoms. This review investigates the mechanisms by which exercise influences myokines, neurotrophic factors, growth factors, and inflammation-related factors to promote neuronal survival and plasticity in Parkinson’s disease. Despite promising findings, the specific molecular pathways through which exercise exerts neuroprotective effects remain largely unexplored, and individual variability in disease progression necessitates personalized exercise interventions tailored to each patient’s needs. Furthermore, for patients unable to engage in physical activity, exploring alternative therapies that mimic exercise to achieve neuroprotective effects is crucial. In conclusion, this review highlights the need for further research to elucidate the molecular mechanisms of exercise-induced neuroprotection and to establish effective individualized exercise programs, ultimately improving the management of Parkinson’s disease.

1 Introduction

Parkinson’s disease (PD) represents a neurodegenerative condition primarily affecting dopaminergic neurons situated within the substantia nigra region of the midbrain. This disorder unfolds through a gradual clinical progression, encompassing a spectrum of symptoms that include premotor, non-motor, and motor manifestations. Collectively, these symptoms profoundly impair patients’ quality of life and contribute to substantial healthcare expenditures (Leite Silva et al., 2023). Simultaneously, caregivers of those diagnosed with PD are under tremendous stress (Macchi et al., 2020). The primary movement-related signs characteristic of PD encompass bradykinesia, muscle tonus, quiescent tremor, and postural instability (Ahmad et al., 2023). Although the etiology of PD continues to be enigmatic, contemporary research indicates that the development of Lewy bodies (Shahmoradian et al., 2019), neuroinflammation (Tansey et al., 2022), mitochondrial damage (Eldeeb et al., 2022), and intestinal flora dysbiosis (Zheng et al., 2021), and other factors that may be closely related to PD.

Age constitutes the primary risk factor associated with the onset of PD (Tolosa et al., 2021). With the ongoing enhancement of survival rates within the elderly population, a corresponding rise has been observed in the total count of individuals diagnosed with PD, a trend that is anticipated to continue. This situation is described by some as a pandemic (Dorsey et al., 2018). There is often a long delay (10 years on average) between the first manifestation of symptoms and the time of diagnosis in people with PD, due to the fact that PD is challenging to identify in its initial phase (Gaenslen et al., 2011). Initial manifestations of the condition include constipation (the most prevalent indication), the occurrence of dream activity occurring during the rapid eye movement (REM) sleep stage (suggestive of REM sleep-behavioral dysfunction), diminished sense of smell, unilateral and indistinct shoulder discomfort, or depressive symptoms (Armstrong and Okun, 2020). None of these early manifestations are sufficient to establish a designation of PD by themselves, as each of them can also occur as a part of many other diseases. As a result, the disease is typically diagnosed late in the course of complete neuronal degeneration, and treatment is always delayed (Lotankar et al., 2017). Taking early preventive measures becomes an effective way to reduce the incidence of PD. It has been increasingly demonstrated that a certain amount of physical activity can prevent neurodegeneration, which is associated with PD (Ritz and Paul, 2022). Under certain assumptions, introducing vigorous physical activity in a population could prevent 14.5% of current PD cases (Ben-Shlomo et al., 2024).

Exercise not only decreases the risk of developing of PD, it is also an effective treatment. For motor symptoms in PD patients, effective therapeutic interventions such as Tai Chi, yoga, or traditional balance exercises can optimize balance and mobility (Zhang T. et al., 2023). Motor deficits of rat models simulating PD are ameliorated after prolonged treadmill routine interventions. Following physical exercise, there is a reduction in oxidative stress levels and an enhancement in mitochondrial function observed in rat models (Landry et al., 2022; Liu L. et al., 2024; Mu et al., 2022; Tung et al., 2024). Expiratory muscle training has the potential to markedly diminish the overall seriousness of dysphagia in individuals suffering from PD (Claus et al., 2021). Meanwhile, physical activity can also affect Parkinson’s-related non-motor manifestations and may represent a viable therapeutic approach for alleviating fatigue among those with PD (Folkerts et al., 2023). Yoga also has a considerable impact on anxiety and depression among patients with PD (Kwok et al., 2019). Furthermore, dance interventions offer both motor and non-motor benefits for Parkinson’s disease (PD). Within the sensorimotor domain, therapeutic effects predominantly manifest in balance improvement, gait enhancement, reduction of motor symptom severity, and increased functional mobility among individuals with PD (Bek et al., 2020). Through structured dance participation, persons with PD experience not only neuromotor improvements but also opportunities for self-actualization and social reintegration, with measurable benefits in combating disease-related stigma and enhancing perceived wellbeing (Bognar et al., 2017). It is shown that physical activity also affects the pathologic manifestations associated with PD in related studies. Aerobic exercise (but not stretching) leads to enhanced connectivity function between the anterior shell region and the sensorimotor cortex area relative to the posterior shell nucleus (Johansson et al., 2022), and exercise on a treadmill alleviates the propagation of α-syn (Dutta et al., 2022). Nonetheless, it is of paramount importance to acknowledge that effective exercise is a significant strategy for reducing the risk of PD and assisting in the treatment of PD as well as reducing its mortality rate, but only with appropriate intensity and duration of exercise (Bhadra et al., 2023; Stein et al., 2021; Tsukita et al., 2022; Zhang X. et al., 2022). Engaging in excessive physical exercise can result in bodily tiredness, and the equilibrium of oxidative/antioxidant systems in the body can be disrupted, which in turn causes damage to the central nervous system (CNS) (Bai et al., 2023).

The favorable outcome of exercise in PD is undisputed, but the mechanisms of action are unclear. These mechanisms may involve neuronal survival and plasticity, neurogenesis, epigenetic modifications, angiogenesis, autophagy, etc (Sujkowski et al., 2022). In numerous studies, the impacts of physical activities on the body’s metabolism have garnered extensive discussion, particularly regarding its promotion or inhibition of the production of related cytokines. This paper will delve into the promoting influence of physical exercise on the production and secretion of cytokines, as well as the intrinsic mechanisms of these factors in alleviating symptoms of PD. We will primarily concentrate on myokines, neurotrophic factors, growth factors, and inflammation-related factors. The intrinsic mechanisms of various exercise-induced factors hold significant research importance, as they could prepare for a novel treatment for PD patients who already experience motor impairments and those who are unable to engage in physical activity.

2 Myokine

Myokines are a class of peptide molecules produced and secreted by skeletal muscles during exercise and are considered one of the key mechanisms underlying the neuroprotective effects of physical activity. These factors exert beneficial effects through various regulatory pathways, including promoting cell survival, neurogenesis, modulation of neuroinflammation, maintenance of proteostasis, resistance to oxidative stress, and protein modification. Recent studies on exercise-induced myokines have opened new avenues for the treatment of neurodegenerative diseases (Lee et al., 2021). To date, only a few myokines have been systematically investigated for their roles in such disorders. In this section, we focus on two well-studied myokines, irisin and cathepsin B (CTSB), and their effects on neurodegenerative diseases. However, further studies are urgently needed to identify more myokines and comprehensively evaluate their therapeutic potential in neurodegenerative conditions.

2.1 Irisin

Irisin is a muscle-derived myokine first identified as an exercise-responsive hormone, acting as a crucial molecular bridge between physical activity and systemic health. Its secretion is induced by physical exercise through the upregulation of PGC1-α, which activates the transcription of the fibronectin type II domain-containing protein 5 (FNDC5) gene. FNDC5 encodes a type I transmembrane protein that undergoes proteolytic cleavage to release a soluble peptide, irisin. Circulating irisin levels are influenced by exercise intensity and correlate with FNDC5 expression in skeletal muscle (Boström et al., 2012; Huh et al., 2012). Although controversy once surrounded its expression in humans—due to the non-canonical ATA start codon in the human FNDC5 gene and the low specificity of commercial antibodies (Albrecht et al., 2015; Raschke et al., 2013)—several mass spectrometry-based studies have successfully validated the presence of irisin in human blood and cerebrospinal fluid (Jedrychowski et al., 2015; Lee et al., 2014; Ruan et al., 2018). Beyond its metabolic roles, irisin has attracted attention for its protective effects across multiple organ systems, including improving liver function (Polyzos et al., 2014), enhancing systemic glucose metabolism (Perakakis et al., 2017), maintaining musculoskeletal homeostasis (Zhao et al., 2024), and suppressing cancer progression (Pinkowska et al., 2021). Notably, irisin also shows promising potential in neurological disorders. FNDC5 is expressed in the brain and involved in neural differentiation and maturation (Baghi et al., 2021). In PD models, FNDC5 expression is downregulated during the acute phase but elevated in the chronic phase, likely reflecting a compensatory response (Tsai et al., 2021). Irisin can cross the blood-brain barrier, and increased peripheral levels are associated with elevated central concentrations. In the brain, irisin regulates neurogenesis, synaptic plasticity, inflammation, and cognitive function (Islam et al., 2021).

Once secreted, irisin circulates and targets tissues to exert its effects. The irisin secretion cycle targets tissues and exerts its physiological effects through the binding to cellular aV family integrin complex receptors, especially aVb5 phosphorylation of the 397 site of Focal Adhesion Kinase (FAK) after ligand binding. It is worth noting that this signal transduction has high specificity. In HEK293T cells expressing integrin aVb5, irisin treatment significantly enhanced the phosphorylation level of FAK; However, similar effects were not observed in cells expressing integrin aVb3 (Kim et al., 2018). The present research found that the interaction between irisin and the aVb5 receptor is facilitated by the motility-inducing molecule Heat Shock Protein 90α (Hsp90α). The activation of the aVb5 receptor demonstrates a strong attraction toward irisin (Mu et al., 2023). It was confirmed that uncut FNDC5 could directly promote the secretion of BDNF and enhance synaptic plasticity by binding to integrin aVb5 receptor on the surface of hippocampal neurons in MPTP induced chronic PD mouse models. Additionally, FNDC5 promotes dopaminergic connectivity from the substantia nigra to the hippocampus by mediating the interaction between integrin aVb5 in hippocampal neurons and CD90 molecules on the dopaminergic terminal membrane. It is notable that FNDC5 directly binds to the receptor without splicing, while irisin binds to the receptor after splicing (Tang C. et al., 2023). Nonetheless, the complete identification of irisin receptors remains an unresolved challenge in the field (Pignataro et al., 2021), while the intracellular signaling pathways of irisin have been largely elucidated. Irisin primarily exerts its biological functions through mitogen-activated protein kinase (MAPK) signaling pathways, with involvement in processes such as neural differentiation, osteoblast proliferation, and osteogenic differentiation (Rabiee et al., 2020). In addition, irisin mediates additional physiological effects through alternative signaling pathways. For instance, irisin can induce white adipose tissue browning via p38 and ERK signaling pathways (Zhang et al., 2014), The ERK1/2 MAPK signaling cascade mediates endothelial cell proliferation processes (Liu et al., 2017). Irisin demonstrates anti-metastatic activity in lung cancer by reversing EMT processes through PI3K/AKT-dependent inhibition of Snail signaling (Shao et al., 2017).

Currently, there are relevant studies that have demonstrated that dietary changes are unable to influence the standards of irisin within the body (Ko et al., 2016); exercise may be the only current method to increase circulating irisin in the body. For example, related studies have shown that despite similar body weights, baseline levels of irisin were observed to exhibit higher levels in individuals who have undergone training compared to those who have not, suggesting that exercise exerts a direct effect on the elevation of irisin (Algul et al., 2017). Performed on different exercise modalities, compared to endurance training alone as well as in comparison to exercise regimens that incorporate both resistance and endurance components, resistance training resulted in a more significant enhancement of the irisin response (Kim H. J. et al., 2015). Furthermore, the intensity of exercise also has an effect on irisin production. In mouse experiments, male subjects were randomly allocated to a sedentary control group (CO), a free-contact running wheel group (RW), and a running group (TM); furthermore, the findings indicated that the concentrations of irisin were increased in the mice belonging to the TM group immediately after acute exercise. However, in the RW group, the PGC1-α/FNDC5/irisin pathway did not respond significantly to mild exercise (Brenmoehl et al., 2014). The experiment demonstrates that moderate to high-intensity exercise increases the expression of PGC1-α, which in turn elevates the levels of FNDC5, subsequently increasing circulating irisin levels (Benedini et al., 2017). However, there are also studies that offer a different view on whether exercise can increase irisin levels: chronic exercise training brings about a notable diminution in the levels of circulating irisin (Qiu et al., 2015), while acute strength training lasting up to 30 min did not cause a significant elevation of serum irisin levels (Pekkala et al., 2013). Although some studies also mentioned a decrease in irisin after exercise, most of the studies on irisin still showed an increase after exercise (Table 1), which could be due to differences in assay method or time of day.

TABLE 1
www.frontiersin.org

Table 1. Comparative analysis of exercise interventions and changes in irisin levels.

Irisin hormone, a myokine, has a vital impact on several neurodegenerative disorders (Ali et al., 2024), and various studies have demonstrated that irisin effectively inhibits the pathologic changes in PD (Figure 1). Neurodegeneration, which is induced by pathological α-synuclein (α-syn), was prevented by irisin in the α-syn preformed fibers (PFF) mouse model of PD, thereby preventing the deterioration of dopaminergic neurons and the subsequent decrease of striatal dopamine (De Sousa, 2024). The intrinsic mechanisms can occur in at least three ways: (1) The coming into being of α-syn phosphorylated at serine 129 and the death of neuronal cells were diminished by irisin. And then irisin conferred neuroprotective effects against the neurotoxicity induced by α-synuclein. (2) The internalization and aggregation of pathological α-syn were reduced by irisin; therefore, irisin could achieve the effect of reducing its accumulation. (3) The efficiency of irisin in enhancing lysosomal degradation-mediated synaptophysin clearance effectively prevented the pathological transmission of α-syn. In this process, it was experimentally demonstrated that, 1 and 4 days after α-syn PFF treatment, irisin reversed 100% and 34.6% of the affected proteins, respectively. Notably, irisin treatment led to a significant upregulation of the ApoE protein (Kam et al., 2022). The ε 4 genotype of ApoE protein regulates α-Syn pathology in humans (Davis et al., 2020) and is linked to a heightened risk of dementia among patients with PD, while ApoE ε 2 may prevent the aggregation of α-syn and its link with neurodegenerative processes in synucleinopathies (Bras et al., 2014; Mata et al., 2014). Other studies have determined by immunoblotting that irisin rescues dopamine transporter protein (DAT)-positive and tyrosine hydroxylase (TH)-positive fibers, prevented the loss of dopamine neurons and lowering of striatal dopamine (Kam et al., 2022), and effectively promotes dopamine absorption in the contralateral striatum of the affected limb in PD patients (Shi et al., 2024). Iron death shares common features with PD pathophysiology (Mahalakshmi et al., 2020). It is reported that irisin can improve the inflammatory microenvironment by inhibiting hippocampal ferroptosis through the Nrf2/GPX4 signaling pathway, significantly decreasing the abundance of ROS, MDA, ASCL4, and ALOX12, and decreasing the progression of neuroinflammation (Wang J. et al., 2022). In the neurotoxicity-induced PD model, irisin does not directly target mitochondria after being transported to cells but activates the signaling pathway through binding to the αV integrin receptor to exert protective effects. Irisin activates the phosphorylation of FAK, Akt, and ERK1/2. Then the signaling processes mediated by PI3K-Akt and ERK1/2 are activated. Next, it refreshes mitochondrial function, restores mitochondrial morphology, and prevents apoptosis and oxidative stress, etc. Besides, irisin can also alleviate the mitochondrial damage through the MAPK signaling pathway. It is noteworthy that during irisin treatment, it bypasses the AP site (DNA damage abasic site) caused by ROS and restores OGG1, the main enzyme responsible for repairing 8-oxoG as well (Zhang X. et al., 2023). While simultaneously, irisin has the capability to stimulate the AMPK signaling pathway within the brain, mitigating the generation of inflammatory cytokines and alleviating neuroinflammation and oxidative stress (Zhang Q. et al., 2024). Various studies have suggested that irisin may play a role in different animal models of PD. However, current research on its underlying mechanisms remains incomplete, and further prospective studies are needed to identify the specific molecular pathways through which irisin counteracts the pathological features of PD.

FIGURE 1
Diagram illustrating the effects of various exercises (rowing, cycling, weightlifting) on muscle and brain health. Exercise activates PGC1-α and FNCD5 in muscles, releasing irisin. Irisin crosses the blood-brain barrier, affecting brain cells by inhibiting hippocampal ferroptosis, reducing ROS and inflammatory factors, and decreasing neuroinflammation. A close-up box shows reduced phosphorylated α-syn levels, reduced accumulation of pathological α-syn, and increased lysosomal degradation. This process involves pathways like Nrf2-GPX4, AMPK, and AKT/ERK, contributing to improved brain health.

Figure 1. Exercise boosts irisin and helps PD treatment. When the human body engages in exercise, muscle cells are stimulated to secrete PGC1-α, which further promotes the expression of FNCD5. The irisin released by FNCD5 can cross the damaged blood–brain barrier and enter the brain, triggering a series of protective mechanisms. In terms of inhibiting neuroinflammation, irisin activates the AMPK signaling pathway, decreases the level of ROS (reactive oxygen species), and increases the expression of OGG1 (8–oxoguanine DNA glycosylase 1). Meanwhile, it activates the Nrf2–GPX4 pathway. Together, they inhibit ferroptosis. In addition, the levels of inflammatory factors decrease, and the AKT/P13k and ERK1/2 signaling pathways are also activated to update mitochondrial function, thereby reducing neuroinflammation. In the aspect of regulating α-synuclein (α-syn) metabolism, irisin reduces the number of phosphorylated serine on α-syn, prevents the aggregation of pathological α-syn, and promotes its degradation by lysosomes, thus alleviating the pathological accumulation of α–syn in the brain.

2.2 CTSB

Within the lysosome, cleavage of the propeptide converts procathepsins into mature cathepsins. Cathepsins are integral to various lysosome-related processes, including protein degradation, metabolism, renewal, antigen presentation, apoptotic signaling, phagocytosis, and growth factor receptor recycling, particularly in autophagy (Turk et al., 2012). Therefore, dysregulated expression or activation of cathepsins is associated with a wide range of autoimmune diseases, phlogosis (Patel et al., 2018), and even neurodegenerative diseases (Stoka et al., 2016). CTSB that encodes cathepsin B is the most prevalent cysteine protease among all lysosomal proteases and is widely expressed (Yadati et al., 2020), especially in the CNS, where it is linked to neuronal function (Tran and Silver, 2021).

Exercise mediates the advantageous consequences of CTSB on brain function through various mechanisms. Running increases the expression of the CTSB gene in the hippocampus. Physical activities can lead to low oxygen level (Radak et al., 2013), which can enhance the quantities of CTSB in the brain (Yakovlev and Gulyaeva, 2015). The removal of neurodegenerative fragments (Devi and Kiran, 2004) and adult neurogenesis can be promoted by this elevation, a process related to memory operations (Abrous and Wojtowicz, 2015). The suppression of CTSB and cathepsin L (CTSL) reduces the synthesis of BDNF in the hippocampus (Bednarski et al., 1998). BDNF engages in the regulation of synaptic plasticity, the survival of cells, and differentiation (Chao et al., 2006).

Genetic differences in CTSB have been proven to be hazardous factors for PD (Pedersen, 2019). A cellular characteristic of PD is the cytoplasmic amassment of α-syn and amyloid formation. CTSB can proteolyze the core and amyloidogenic regions of α-syn, thereby preventing the taking shape of α-syn fibrils (McGlinchey and Lee, 2015). There may be crosstalk between α-syn aggregation and lysosomal-autophagy damage (Bellomo et al., 2020). When α-syn aggregates, the transport of CTSB to lysosomes is disrupted, resulting in that proteolytic activity of cathepsins decreased. Thus, promoting the transport of CTSB to lysosomes can increase their activity, facilitating efficient degradation of α-syn (Drobny et al., 2023). In summary, exercise-mediated elevation of CTSB demonstrates its benefits for PD and highlights the importance of CTSB in degrading α-syn, highlighting its latent as a treatment mark for PD.

3 Neurotrophic factors

Neurotrophic factors are a group of secreted proteins that support neurogenesis, promote synaptic plasticity, and prevent neuronal degeneration under both physiological and pathological conditions. They play a crucial role in the survival, maintenance, and regeneration of specific neuronal populations in the adult brain (Allen et al., 2013). Physical exercise has emerged as an effective stimulus for the expression of neurotrophic factors, thereby contributing to neuroprotection in neurodegenerative diseases (Wang and Holsinger, 2018). Key neurotrophic factors influenced by physical activity include brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). In this section, we summarize the regulation of exercise-induced neurotrophic factors and their potential therapeutic roles in neurodegenerative diseases.

3.1 BDNF

BDNF, it is regarded as the most plentiful neurotrophic factor in the brain of adults, with a role as a significant neurotrophic signal and neuromodulator (Bazzari and Bazzari, 2022). In the brain, BDNF is expressed by glutamatergic neurons (Andreska et al., 2014), glial cells (e.g., astrocytes isolated from the cortex and hippocampus but not from the striatum) (Clarke et al., 2018), and microglia (Parkhurst et al., 2013). It is found extensively in the CNS and PNS (peripheral nervous system), as well as in bone and cartilage tissues, and the highest concentration was located in the brain and cortex. The BDNF molecule consists of three structural domains: a signal peptide, a pre-structural domain, and a mature structural domain (Colucci-D’Amato et al., 2020). After entering the Golgi apparatus, the signal peptide is cleaved off. BDNF is first produced as the proBDNF precursor molecule; then, the proBDNF molecule can be cleaved intracellularly by furin or extracellularly by fibrinolytic enzymes or matrix metalloproteinases to form BDNF (Figure 2; Finan et al., 2018). Furin identified as the first proprotein convertase discovered in mammals (Thomas, 2002), cleaves proprotein substrates in the brain, including the precursor of BDNF (Zhang Y. et al., 2022). It plays a protective role in CNS injury in the brain mainly through binding to its specific receptor, the tropomyosin-related kinase receptor B (TrkB). The BDNF-activated TrkB signaling mechanism is critical for the survival of neurons, structural modifications, and plasticity (Wang et al., 2024).

FIGURE 2
Diagram illustrating the role of BDNF (brain-derived neurotrophic factor) in brain health. The left section shows BDNF production through exercise and its processing by furin and fibrinolysin. The right section details BDNF’s effects, including neurogenesis, inflammation, and cognitive functioning, through various pathways like TrkB, CREB, STAT3, and Akt/mTOR. Connections show relationships to apoptosis, cell damage, and neurodegeneration.

Figure 2. The link among exercise, Brain-derived neurotrophic factor (BDNF) and neuroprotection. Exercise enhances both central and peripheral expression of BDNF, providing neuroprotection through multiple pathways.

Research has demonstrated that self-directed physical activity boosts the hippocampal expression of BDNF (Loprinzi et al., 2019), which in turn affects hippocampal neurogenesis (Fabel et al., 2009). Physical activity is known to increase circulating levels of BDNF (Figure 2; Knaepen et al., 2010; Szuhany et al., 2015), thereby improving brain function (Tuon et al., 2015; van Praag et al., 2005; Vilela et al., 2017). Circulating BDNF is present in cerebrospinal fluid and blood, and it has been demonstrated that circulating BDNF in plasma can cross the blood-brain barrier to some extent (Pan et al., 1998). The animal data also show a correlation between peripheral and central concentrations (Klein et al., 2011). Currently, the relationship between exercise and peripheral circulating BDNF has been well studied. A randomized controlled trial (RCT) consisting of 18 studies evaluated the effect of exercise intervention on plasma BDNF levels in patients with neurodegenerative diseases [i.e., multiple sclerosis, Parkinson’s disease, mild cognitive impairment (MCI), and Alzheimer’s disease] compared to a lack of exercise. Overall, the exercise intervention resulted in significantly higher plasma BDNF levels in neurodegenerative diseases (Ruiz-González et al., 2021). However, various forms of exercise impact BDNF in distinct ways. Several studies have indicated that after several weeks of resistance training, the amount of circulating BDNF rises and is independent of the type of resistance training (Yarrow et al., 2010). However, strength training does not appear effective in increasing BDNF (Huang et al., 2014). It has been established through research that aerobic exercises are effective in enhancing BDNF levels (Cassilhas et al., 2012). A meta-analysis including 910 participants (61.3% male, mean age: 42.2 years) revealed that blood concentrations of BDNF were dramatically increased by aerobic exercise rather than resistance exercise training (Dinoff et al., 2016). New research has once again found no major difference between aerobic and resistance training but rather a positive correlation with the duration of exercise (Dinoff et al., 2017). It is thought that BDNF levels increase after several weeks of regular repetitive training (Church et al., 2016). In addition, Nordic Walking has been proven by scholars to increase the level of BDNF. For patients with Parkinson’s disease of varied durations and severities, it represents a safe, feasible, and sustainable aerobic exercise modality (Harro et al., 2022). But it has also been suggested that interval sprint training is more likely to trigger BDNF than steady-state training at a moderate intensity and steady-state training at a high intensity (Reycraft et al., 2020). Although it is unclear which type of exercise maximizes BDNF, all types of exercise certainly have a function in the elevation of BDNF. The mechanisms behind this process by which exercise promotes BDNF production are not well studied, but some studies have suggested that it may be peripheral lactate levels that promote BDNF production (El Hayek et al., 2019), while others propose that exercise influences gut microbiota, indirectly promoting BDNF elevation (Molska et al., 2024). Additionally, some authors suggest that physical activity increases adrenergic (norepinephrine) or serotonergic (5-HT) signaling, thereby promoting BDNF expression (McMorris, 2016).

After binding to TrkB, BDNF activates intracellular signaling pathways, playing crucial roles in maintaining neuronal growth and survival. During the development of embryonic cells, BDNF-TrkB signaling promotes the development of cortical progenitor cells, leading to their maturation into neurons, i.e., neurogenesis (Fabel et al., 2009). It has been shown that BDNF-TrkB-CREB signaling in excitatory cortical neurons of layer V of the cortex mediates the control of slow wave activity (SWA) during sleep, which in turn affects synaptic strength (ElGrawani et al., 2024). Chen et al. (2023) suggested that hippocampal neurogenesis in neonatal HIBD (hypoxic-ischemic brain damage) rats can improve cognitive impairments by promoting the integrated signaling mechanism involving BDNF, its receptor TrkB, and CREB. Numerous clinical studies have found that PD patients have significantly reduced BDNF (Howells et al., 2000; Kang et al., 2017; Karakasis et al., 2011). Inhibiting BDNF expression in the substantia nigra (SN) leads to a PD-like phenotype in rats (Hernandez-Chan et al., 2015). In addition to this, BDNF variants can significantly heighten the possibility of LRRK2 induction among individuals who experienced PD at a certain age greater than 60 years, suggesting that there is a cumulative effect between the two genetic variants and that the simultaneous presence of both increases the likelihood of the development of PD (An et al., 2008), and variants at the LRRK2 locus are a significant genetic risk factor in the development of PD (Liu and LeRoith, 1999). The study of the LRRK2 gene began with the observation of an autosomal dominant inherited Parkinson’s disease case in a Japanese family in 1978. Subsequently, in 2002, the PARK8 locus was identified through linkage analysis, and the p.I2020T mutation was identified (Sosero and Gan-Or, 2023). Thereafter, other LRRK2 variants associated with Parkinson’s disease were identified (Ross et al., 2011). LRRK2 mainly causes harm to dopamine neurons through multiple mechanisms such as inflammatory response (Cook et al., 2017; Rui et al., 2018), lysosomal dysfunction (Eguchi et al., 2018), and mitochondrial dysfunction (Ludtmann et al., 2019), which suggests an important link between PD and BDNF. There are also many studies on the therapeutic effects of BDNF on PD. It has been demonstrated that BDNF in the dorsal striatum (dStr) increases after physical activities, which facilitates an augmentation in the secretion of dopamine in the dStr (Bastioli et al., 2022). Geng et al. (2023) showed a significant decrease in the levels of p-STAT3, p-PI3K, p-AKT, and p-mTOR in a PD mouse model by protein blotting and also suggested BDNF inhibited apoptosis induced by MPP+ and cell proliferation, which was inhibited by siSTAT3 by CCK-8 and flow cytometry. BDNF enhances the phosphorylation of STAT3, which then activates the PI3K/AKT/mTOR signaling axis, regulating neuronal autophagy, preventing apoptosis, and encouraging cell growth. Meanwhile, their study also found that the BDNF group exhibited a notably decreased concentration of p-α-syn protein when compared to the MPP+ group, suggesting that BDNF protects neuronal cells by boosting autophagy and reducing the expression levels of α-syn. In this way, BDNF attenuated cell injury in the PD model. In astrocytes, BDNF-mediated activation of Nrf2 controls circadian rhythms, safeguarding dopaminergic neurons against ferroptosis (Ishii et al., 2019), thereby reducing the degenerative effects that PD may induce. BDNF also has a part in protecting against dopaminergic neurodegeneration in PD by inhibiting neuroinflammation and apoptosis, mainly targeting the pathways of NRF2 and NF-κB signaling interactions (Thirupathi et al., 2024). The potential value of BDNF as a treatment for PD is irrefutable, but it has also been shown that neither the direct delivery of peripheral BDNF to the brain nor promotion of the expression of BDNF gene alleviates the symptoms of PD, and that only physical activity that boosts the levels of BDNF in the brain is effective (Palasz et al., 2020). Although there have been many studies on the mechanism of BDNF in PD (Figure 2), we still need further research to explore the intrinsic mechanisms by which exercise promotes BDNF.

3.2 GDNF

GDNF is a homodimer that is glycosylated and contains disulfide bonds (Lin et al., 1993). It was initially extracted from cultured B49 rat glial cells (Lin et al., 1994). GDNF is distributed in both PNS and CNS, synthesized and released by various cells, including astrocytes, oligodendrocytes, and Schwann cells, and other multiple glial cell types (Sariola and Saarma, 2003).

Nowadays, there is sufficient evidence that exercise promotes GDNF production. BDNF and GDNF are elevated in the flounder muscle, after performing a round of exercise (Dupont-Versteegden et al., 2004; Koeda et al., 2024). Elevated levels of GDNF have been observed during treadmill training (Côté et al., 2011). Voluntary running triggers endogenous GDNF expression (Bonafina et al., 2019). Meanwhile, researchers demonstrated that swimming or running for two weeks changed the GDNF protein levels found in the lumbar spine and muscle tissues of rats that are young and old (de Azambuja et al., 2018; Gyorkos et al., 2014). Regarding the influence of different physical activity modalities on the promotion of GDNF, it has been demonstrated that various training modalities in young animals upregulate GDNF expression, but there are no notable distinctions between exercise modalities (McCullough et al., 2013). Physical training, which takes place both at the beginning and in subsequent stages, rescued nearly all dopaminergic neurons in the substantia nigra and ventral tegmental area, inhibited inflammatory responses, and raised levels of BDNF and GDNF to a comparable degree (Palasz et al., 2019). This could mean that exercise increases both GDNF and BDNF in PD and may have a combined effect. In Rabelo et al.’s (2017) study, the HP (high performance) group ran for 129 min per day, while the SP (standard performance) and LP (low performance) groups ran for 64 and 29 min per day, and although GDNF was elevated in all three groups, GDNF was significantly elevated in the LP group in comparison to the other two groups. This suggests that moderate training may be more effective than high-intensity training in promoting GDNF levels, although the optimal exercise intensity for enhancing GDNF remains to be determined.

GDNF is a promising and significant therapeutic factor for PD (Figure 3). Studies indicate that dopaminergic neurons in primary cultures undergo survival and differentiation processes. GDNF improved these processes by promoting dopamine uptake and enhancing the capacity for high-affinity dopamine absorption, thus enhancing midbrain dopaminergic neurons’ ability to survive in mammals (Lin et al., 1993). The decrease of GDNF in the prefrontal cortex leads to synaptic connections and neural circuit degeneration, contributing to cognitive impairment in PD patients (Tang C. et al., 2023). In a 6-hydroxydopamine model of PD, forced exercise reduces the destruction of dopamine neurons by 6-hydroxydopamine, and further studies have found that this protective effect is partially due to increased availability of GDNF (Smith and Zigmond, 2003). Utilizing novel nanostructured delivery systems to increase GDNF expression can alleviate some cell-related symptoms related to PD observed in laboratory settings (Guzmán-Sastoque et al., 2024). The application of GDNF promotes better connectivity between transplanted dopamine neurons and the striatum and other targets typically supplied by natural dopamine neurons, thereby increasing dopamine levels in the striatum and restoring motor function (Moriarty et al., 2022). One team investigated the effects GDNF has on the survival of dopaminergic neuron and the transmission of dopamine at their terminals, revealing that a lack of GDNF negatively affects dopamine transmission in the prefrontal cortex (Tang C. X. et al., 2023). Moreover, that the dopaminergic protective effects it promotes are associated with responsive regulation of microglia. However, endogenous GDNF is not sufficient to protect dopaminergic neurons from inflammatory damage (Mendes-Oliveira et al., 2023). GDNF protects, restores, and enhances dopaminergic function of the pathway connecting nigra to striatum in a short-term animal model of PD (Mätlik et al., 2022). RET (rearranged in transfection) was identified as an oncogene activated by the recombination of DNA (Takahashi et al., 1985). RET is indispensable for the development of both the peripheral and central nervous systems (Mahato and Sidorova, 2020). Nurr1 is an important transcription factor regulating RET expression. Although accumulation of α-synuclein leads to down-regulation of Nurr1 and consequent attenuation of RET signaling, GDNF/NRTN therapy has the potential to restore this pathway through an “augmentation mechanism,” especially in neurons that retain some RET expression and thus up-regulate RET expression (Björklund, 2021). At the same time, GDNF/RET signaling may be a factor in Akt activation via a Src-dependent non-classical pathway that degrades α-nucleosynaptic proteins (Chmielarz et al., 2020).

FIGURE 3
Diagram illustrating the interactions of three growth factors: IGF-1, VEGF, and NGF, and their effects on neurons. The IGF-1 section highlights survival of apoptotic neurons, influenced by estrogen and resistance training. VEGF is linked with lactic acid impacts and neuron degeneration. NGF emphasizes neurogenesis and motor function. Pathways involve various proteins and conditions affecting neuronal health and therapy options, such as exercise and stem cell intervention.

Figure 3. Exercise promotes growth factors for better treatment of PD. Exercise increases growth factors—IGF-1, VEGF, NGF. The elevated IGF level, after binding to IGF-1R, promotes the protection of dopaminergic neurons in the SNpc through the PI3K/Akt pathway by estrogen. VEGF and NGF through bone marrow derived stroma cell (BMSC), cAMP responsive element-binding protein (CREB) and other pathway receptors, promotes the survival of dopaminergic neurons and inhibits the progression of neurodegenerative diseases.

Research shows GDNF-transfected macrophages effectively treat Parkin Q311X(A) transgenic mice which are characterized by slow progression and mild cerebral inflammation (Zhao et al., 2019). In late stages, they restore motor function and preserve brain tissue. Early injection in PD mice offers long-lasting benefits, reducing inflammation, α-synuclein buildup, and improving neuron survival and motor function. Thus, they’re promising for advanced and early PD. In conclusion, GDNF-transfected macrophages represent a promising therapeutic strategy for advanced diseases, including the early onset of PD (Zhao et al., 2019). In various clinical trials involving GDNF, PET imaging had indicated an increase in 18F-DOPA uptake. And there was no notable enhancement in the motor function of participants in placebo-controlled studies, which may be due to a lack of improvement attributed to the ineffectiveness of GDNF in enhancing the condition of damaged dopamine neurons (Barker et al., 2020), possibly suggesting that GDNF acts mainly during the initial phases of PD or primarily makes a preventative contribution. However, more GDNF is not necessarily better. Overexpression of GDNF can downregulate levels of tyrosine hydroxylase (TH), a key enzyme in dopamine synthesis, in rats, and may lead to abnormal sprouting and ectopic formation of synapses in the brain, resulting in various dose-dependent adverse effects (Georgievska et al., 2004).

4 Growth factors

Growth factors are a class of cytokines with the ability to stimulate cell growth. Most of them are proteins or polypeptides that can regulate key cellular activities such as growth, proliferation, and differentiation, playing crucial roles in human development, tissue repair, and immune regulation (Wang, 2021). Growth factors can protect and restore degenerated neurons while enhancing their functional activity, thus holding considerable promise for improving neurodegenerative diseases (Sidorova and Saarma, 2020). Exercise therapy can promote the production of growth factors, thereby facilitating the prevention and treatment of Parkinson’s disease (Pahlavani, 2023). Therefore, in this section, we will focus on discussing the impact of exercise on the production of IGF-1, VEGF, and NGF, as well as how these three growth factors exert neuroprotective effects.

4.1 IGF-1

A hormone called insulin-like growth factor 1 (IGF-1) is released by the liver cells, with its production increasing in the presence of growth hormone. IGF-1 stimulates myelination, oligodendrocyte maturation, neurite outgrowth, and neuronal survival in the brain (D’Ercole et al., 1996).

The effects of exercise on IGF-1 vary under different conditions, including the exercise population and type. Resistance training (RT), aerobic training (AT), or combination training (CT) raised IGF-1 levels in elderly individuals suffering from sarcopenic obesity, according to an experiment that looked at the effects of various forms of exercise on body composition, muscle strength, and IGF-1 levels. However, the effects of different exercise modalities on IGF-1 levels varied, with resistance training showing the most significant effect overall (Chen et al., 2017; Feng et al., 2022). This finding is also supported by other studies. The effects of several training modalities has been described on skeletal muscle mass, function, and IGF-1 signaling in aged mice in their research, Li et al. (2022) found that resistance training was superior to other forms of exercise in raising IGF-1 levels. Exercise has different impacts on IGF-1 levels in the short and long term. Stein et al. found that following acute resistance exercise, Alzheimer’s disease (AD) patients exhibited higher circulating IGF-1 levels compared to healthy individuals (Stein et al., 2021). Additionally, the duration and intensity of exercise significantly affect IGF-1 levels. One study discovered that after a single 30-min exercise session, IGF-1 levels in healthy older males increased; however, after 12 weeks of training, IGF-1 levels decreased (Behrendt et al., 2021). In a study of 14 healthy middle-aged males who participated in a 100-kilometer walking marathon, researchers observed a decrease in IGF-1 levels following high-intensity walking (Kim T. et al., 2015). This phenomenon is not only observed in humans but also animal models. In an experiment involving 20 male Wistar rats to investigate IGF-1 gene expression in colon mucosa with varying exercise levels, Buehlmeyer et al. (2007) found that serum IGF-1 levels decreased as exercise intensity increased. This reduction may be due to high-intensity exercise activating p53, leading to increased expression of p21, IGFBP-3, and PTEN, which induces negative regulation of the IGF-1 pathway (Yu et al., 2016). Notably, teenagers’ resting blood IGF-1 levels throughout brain development are positively impacted by prolonged, consistent aerobic activity (Jeon and Ha, 2015). Therefore, selecting an appropriately regular exercise regimen is essential to enhance serum IGF-1 levels while avoiding reductions associated with excessive exercise.

Male sex was found to be the highest-ranking risk factor, closely followed by serum IGF-1, when the relative impacts of non-genetic variables in Parkinson’s patients were examined using a comprehensive machine learning algorithm (IDEARS) (Allwright et al., 2023). Serum levels of IGF-1 were higher in Parkinson’s patients than in the normal group, with early-stage patients showing higher levels than those in mid-to late-stage disease, according to enzyme-linked immunosorbent assays conducted on serum IGF-1 levels in 100 healthy controls and 100 PD patients (including 49 early-stage and 51 mid- to late-stage cases). This may indicate that IGF-1 acts as a neuroprotective factor, compensatorily elevating in early PD to protect dopaminergic neurons from degeneration (Godau et al., 2010). Furthermore, in patients with PD, non-motor symptoms like anxiety, depression, and cognitive dysfunction were substantially inversely connected with serum IGF levels (Shi et al., 2023). Silencing information regulator 2 related enzyme 1 (sirtuin1, SIRT1) is an NAD-dependent deacetylase, which participates in a multitude of biological processes, including cell cycle regulation, DNA repair, apoptosis and inflammation, autophagy, and aging, and plays a pivotal role in preventing neurodegenerative diseases (Chen C. et al., 2020; Lamichane et al., 2019; Xie et al., 2019). SIRT1 silencing extended the survival of neurons with apoptotic damage by inducing IGF-1 protein expression and secretion, as well as increasing IGF-1 receptor (IGF-1R) protein levels (Sansone et al., 2013). Blocking central IGF-1 receptors (IGF-1R) can weaken estrogen’s neuroprotective effects on SNpc dopaminergic neurons. Additionally, the PI3K/Akt signaling pathway, not the MAPK/ERK pathway, is required to safeguard SNpc dopaminergic neurons by both estrogen and IGF-1 (Quesada et al., 2008). In a rat model of PD, human neural progenitor cells that overexpressed IGF-1 also preserved dopaminergic neurons and recovered their functionality (Ebert et al., 2008). The protective effect of IGF-1 on PD is undeniable (Figure 3), and it may also serve as an early biomarker for PD.

4.2 VEGF

Vascular endothelial growth factors (VEGFs) are members of the VEGF/PDGF (platelet-derived growth factor) group, which is a subfamily of the cystine knot superfamily of hormones and extracellular signaling molecules (Vitt et al., 2001), and are classified as secretory polypeptides. As a component of the PNS, the enteric nervous system (ENS) functions primarily independently while maintaining a connection to the CNS through the gut-brain axis. Hecking et al. (2022) provided experimental evidence of VEGF’s direct neuroprotective effects in the ENS. VEGF plays a significant and potent neuroprotective role following various types of central nervous system damage, such as epilepsy (Nicoletti et al., 2008), cerebral ischemia (Guo et al., 2016), oxidative stress (Cabezas et al., 2019), and neurodegenerative diseases (Guo et al., 2019; Lange et al., 2016). Calvo et al. (2022) found that a lack of retrograde transport of VEGF from the periphery affects the physiology of intact motoneurons, leading to a state akin to axotomy. This implies that VEGF is a necessary retrograde factor for motoneurons’ synaptic drive and discharge activity.

Exercise’s impact on VEGF has been the subject of numerous research studies. One study demonstrated that after eight weeks of moderate-intensity running, VEGF mRNA expression in rat skeletal muscle increased (Shin et al., 2015). Exercise raises VEGF levels through lactate generation, as evidenced by the fact that high-intensity interval training has been demonstrated to enhance VEGF-A levels in the brains of wild-type rats but not HCAR1 knockout mice (Morland et al., 2017). Zang et al. (2023) stated that exercise enhances the expression of the proangiogenic factor VEGF while reducing the expression of the antiangiogenic factor endostatin. This regulation occurs through a partially nitric oxide (NO)-dependent mechanism, promoting capillary growth in the cerebral cortex and improving cognitive function. Different types of exercise have different impacts on VEGF levels. It is noteworthy that VEGF promotes abnormal angiogenesis; therefore, anti-VEGF therapy is necessary in tumors and ocular diseases (Mabeta and Steenkamp, 2022; Mettu et al., 2021). Meanwhile, VEGF serves as a crucial neurotrophic factor for motor neurons, holding significant therapeutic potential in treating motor neuron diseases (Calvo et al., 2024). This underscores the context-dependent nature of VEGF’s effects, which cannot be reduced to a simplistic binary of “beneficial” or “harmful” but rather depends on its site of action, concentration levels, and surrounding microenvironment. Notably, exercise-induced VEGF upregulation typically represents a physiological, controlled, and mild elevation. At the conclusion of exercise, VEGF is increased by 128% ± 36% from resting levels when sprint interval training (SIT) is performed (p = 0.017). Conversely, other forms of exercise, like continuous moderate-intensity exercise and high-intensity interval training based on guidelines, do not exhibit comparable noteworthy gains (Weaver et al., 2021). Research indicates that acute endurance exercise elevates VEGF expression more intensely than strength training, while long-term endurance training enhances an individual’s sensitivity to VEGF responses during any exercise. Therefore, the exercise pattern and endurance training status significantly differ in their effects and mechanisms for increasing VEGF expression (Bizjak et al., 2021). Three hours post-exercise, long sprint interval training (LSI) and moderate-intensity continuous training (MIC) show significantly higher VEGF mRNA increases compared to short-duration high-intensity sprints (SSI), indicating that prolonged high-intensity interval training and moderate continuous training are more effective at activating VEGF gene expression than brief high-intensity sprints (Fiorenza et al., 2020). These results imply that exercise promotes VEGF production, regulates the balance of angiogenesis in the brain, and enhances vascular expansion (Soori et al., 2022).

VEGF supports the neuroprotection of dopaminergic neurons through both indirect and direct mechanisms, inhibiting the apoptosis of dopaminergic neurons in vitro (Mihci et al., 2011). In a mouse model of 6-hydroxydopamine (6-OHDA) damage, the injection of mesenchymal stem cells expressing VEGF189 significantly alleviates mechanical allodynia in PD mice, achieved by downregulating TRPV1 in the spinal dorsal horn (Li et al., 2023). VEGF treatment dramatically alleviated motor function impairments caused by 6-OHDA and prevented loss of dopaminergic neurons in the pars compacta of the substantia nigra (SNpc) and dopaminergic fibers in the striatum, according to another study that used a unidirectional 6-OHDA PD model. Furthermore, in the rat PD model, VEGF gene delivery inhibited microglial activation and apoptosis (Sheikh et al., 2017). In experiments involving stem cell therapy for PD, VEGF promotes local angiogenesis in brain tissue, potentially enhancing the survival rate of transplanted cells, and may also directly protect dopaminergic neurons, reducing their degeneration (Daviaud et al., 2015). Furthermore, there is a synergistic effect between VEGF and GDNF, aiding in the restoration of the dopaminergic system’s topological distribution in severe PD models and alleviating the selective neuronal vulnerability induced by 6-OHDA (Requejo et al., 2017). However, some teams suggest that oligomeric α-synuclein mediates the role of astrocyte-derived VEGFA in the disruption of the BBB associated with PD (Lan et al., 2022). Thus, the connection between PD and VEGF cannot be simply categorized as beneficial or harmful (Figure 3); its effects may vary at different sites or stages of PD progression, warranting further investigation.

4.3 NGF

Levi-Montalcini (1987) discovered and examined nerve growth factor (NGF), the first member of the neurotrophin (NT) family, at the beginning of the 1950s. NGF is a protein with a key role in the growth and differentiation of immature sympathetic nerve cells and in the maintenance of fully differentiated sympathetic neurons (Calissano and Levi-Montalcini, 1979). The cortex, hippocampus, and pituitary gland produce the most NGF, while the basal ganglia, thalamus, spinal cord, and retina also produce sizable amounts (McAllister, 2001). NGF is strongly related to the management of PD and is a crucial regulator of neuronal survival, development, and function. Its roles include regulating neuronal growth, proliferation, activation, and survival (Maes et al., 2023), modulating perception (Testa et al., 2021), maintaining cholinergic neurons (Bruno and Cuello, 2006), and influencing synaptic plasticity in limbic regions, thereby affecting learning and memory capabilities (Ciafrè et al., 2020). However, the mechanisms by which NGF elevation during exercise improves PD remain to be further explored.

Research indicates that NGF levels significantly increase following unilateral eccentric exercise, potentially linked to the onset of delayed onset muscle soreness (DOMS) (Koeda et al., 2024). Progressive exercise also notably raises NGF levels in the PD rats’ striatum (Fallah Mohammadi et al., 2019). After eight weeks of running training in rodents, NGF and its receptor, tropomyosin receptor kinase A (TrkA), are upregulated in the hippocampus (Hong et al., 2015). Cyclic AMP response element-binding protein (CREB) is activated when NGF binds to TrkA, promoting neuroplasticity and cell survival (Lin et al., 2018). Regular resistance training effectively reverses age-related declines in NGF levels (Rahmati et al., 2022). A study found that after four weeks of exercise, proNGF levels in the saliva of both adults and children decreased; particularly, this decrease was linked to increases in creativity and cognitive flexibility (Venditti et al., 2015), which may be because NGF is consumed after it is produced.

NGF is essential for neuron survival and differentiation and has garnered significant attention in neurodegenerative disease research (Figure 3). For instance, PC12 cells differentiated by NGF exhibit enhanced neurotransmitter secretion levels, reduced growth rates, and increased dendritic number and length closely resembling neurons. Thus, PC12 cells are considered an ideal cell line for modeling PD (Zhong et al., 2024). In experimental PD animal models, transgenic bone marrow stromal cells (BMSC-NGF) that produce NGF can be injected into the striatum to promote neurogenesis and markedly enhance motor capabilities (Wang et al., 2008). A reduction in NGF levels is commonly observed in PD, while PI3K/Akt, ERK1/2, and CREB signaling pathway activation may counteract this decline, thereby rescuing damaged neurons (Luo et al., 2018). Additionally, the intracerebral injection of AAV2-NGF has shown good tolerance and therapeutic effects on cognitive decline (Chen W. et al., 2020).

Many studies have shown that exercise significantly affects neurotrophic factors and growth factors (Table 2). However, there is currently no conclusive evidence indicating whether a synergistic interaction exists between the two in this process, which warrants further research.

TABLE 2
www.frontiersin.org

Table 2. Comparative analysis of exercise interventions and changes in neurotrophic factor and growth factor levels.

5 Inflammatory factor

The pathological characteristics and indications for PD are significantly influenced by inflammation. PD subjects’ brains, cerebral spinal fluid (CSF), and blood are known to contain elevated levels of a variety of inflammatory chemicals (Chen et al., 2018; Qin et al., 2016). Tumor necrosis factor (TNF) and interleukins are two examples of inflammatory indicators that are important signaling molecules of immunological activation that affect the peripheral systems and the brain (Tabas and Glass, 2013). While certain cytokines, chemokines, and other inflammatory indicators in PD patients’ peripheral blood exhibit irregularities, dysregulated variables such as IL-6, TNF, IL-2, IL-10, CRP, and CCL5 are frequently found (Qin et al., 2016; Qu et al., 2023), reinforcing the evidence of inflammation-related clinical manifestations in PD. Some scholars have proposed that under pathological conditions, the blood-brain barrier undergoes alterations, allowing the infiltration of peripheral immune cells and inflammatory factors. This leads to a reduction in neuroplasticity-related molecules such as BDNF and GDNF, further weakening the adaptive capacity of the central nervous system (Policastro et al., 2020; Patterson, 2015).

Exercise interventions show promise in enhancing brain plasticity and reducing neuroinflammation (Gupta et al., 2024; Liu Y. et al., 2024). Pro-inflammatory cytokines linked to low-grade chronic inflammation, including IL-6, TNF-α, and monocyte chemoattractant protein-1 (MCP-1), have been shown to be reduced by regular exercise (Martínez-Guardado et al., 2022). Exercise on a rotarod successfully reduced microglial activation and the expression of pro-inflammatory markers such as p-IκBα, iNOS, TNF-α, and cathepsin D in an MPTP mouse model, while raising the expression of anti-inflammatory IL-10 and TGF-β (Leem et al., 2023). Exercise significantly ameliorates inflammatory factors, especially IL-6, and TNF-α (Zhang S. et al., 2024). There are also a large number of experiments to prove this idea (Table 3). Therefore, we will focus on introducing these two inflammation-related factors: IL-6, and TNF-α.

TABLE 3
www.frontiersin.org

Table 3. Comparative analysis of exercise interventions and changes in inflammatory factor levels.

5.1 TNF-α

Carswell et al. (1975) experimentally proved that a protein known as “tumor necrosis factor” (TNF) enters an animal’s bloodstream after lipopolysaccharide assault and reticuloendothelial system stimulation. Activated mononuclear macrophage cells produce the homotrimeric protein known as tumor necrosis factor alpha (TNF-α), which consists of 157 amino acids (Horiuchi et al., 2010). It was the first identified factor with anti-tumor activity (Moore et al., 1999) and is also recognized as a systemic inflammatory response mediator (Blum and Miller, 1998). When activated microglia signal through TNF-α in response to aging, injury, or neuroinflammation, a neurotoxic astrocyte phenotype known as A1, A1-like, or neuroinflammatory reactive astrocytes is induced. These reactive astrocytes suppress supportive functions and start releasing chemokines like CXCL10, complement components like C3, and neurotoxic factors. These effects may help attract immune cells into the CNS and promote inflammation by allowing them to pass through the BBB (Lawrence et al., 2023). Research has indicated that PD patients had higher levels of TNF-α (Liu et al., 2022), underscoring the importance of investigating the intrinsic mechanisms of TNF-α in PD.

The role of TNF-α in PD is dual-faceted (Figure 4). On one hand, TNF-α is a key mediator of neuroinflammation in PD, and its prolonged release can activate microglia and astrocytes, resulting in neurodegeneration and neuronal death (Amin et al., 2022; Shastri et al., 2013). In chronic PD macaque models, serum and CNS levels of TNF-γ and IFN-α were found to be consistently high, synergistically triggering glial activation within the substantia nigra (Barcia et al., 2011). On the other hand, TNF-α may exhibit neuroprotective properties under certain conditions, such as inducing unconjugated bilirubin (UCB) to counteract dopamine damage (Jayanti et al., 2022). Additionally, for the preclinical stage of PD, TNF-α is seen as a potential risk biomarker (Majbour et al., 2020), with plasma levels closely linked to elevated phosphorylated α-synuclein (p-α-syn), which is significantly connected to fatigue symptoms in PD patients, implying its possible involvement in distinguishing fatigued from non-fatigued PD patients (Wang et al., 2023). Interestingly, research in PD mouse models shows that gut microbiota can cause motor impairment by activating the TLR4/TNF-α signaling pathway, but fecal microbiota transplantation (FMT) can reduce symptoms by preventing neuroinflammation and TNF-α signaling (Sun et al., 2018). Moreover, TNF-α promotes the secretion of α-synuclein via lysosomal exocytosis (Bae et al., 2022) and exhibits complex interactions with α-synuclein fibrils during neuroinflammation; when both are present, TNF-α-induced pro-inflammatory responses are partially suppressed (Russ et al., 2021). In neurodegenerative illnesses, TNF-α plays a crucial role, as evidenced by the substantial correlation between high levels and the late course of PD (Piri et al., 2022).

FIGURE 4
Diagram illustrating the impact of NF-kB inflammatory cytokines TNF-α and IL-6. TNF-α leads to glial activation, lysosomal exocytosis, and neurogenic inflammation, affecting dopamine neurons and α-synuclein levels. IL-6 causes dopaminergic neuron degeneration, ASIC1a activation, iron accumulation, and ferroptosis. Exercise has an inhibitory effect on these pathways.

Figure 4. Exercise is anti-inflammatory and protects the brain. The Toll-like receptors (TLRs) on the surface of microglia and the nuclear factor kappa light-chain enhancer of activated B cells (NF-κB) is activated, thereby triggering the formation of inflammasomes and increasing inflammatory cytokines such as tumor necrosis factor (TNF), and interleukin-6 (IL-6). An experiment found that the elevation of IL-6 leads to dopaminergic degeneration. Exercise can reduce the neural damage caused by inflammatory factors and improve cognition.

5.2 IL-6

In 1973, interleukin-6 (IL-6) was first identified as a soluble factor secreted by T cells that is essential for antibody production by B cells (Hirano et al., 1986; Kishimoto and Ishizaka, 1973). IL-6 is a multifunctional cytokine that exerts both pro-inflammatory and anti-inflammatory effects (Scheller et al., 2011). IL-6 activates Janus kinases (JAK) by signaling either the trans-signaling pathway (sIL-6R) or the classical signaling pathway (mIL-6R) (Li et al., 2014), which in turn initiate three possible signaling pathways (Kaur et al., 2020). The first involves the phosphorylation of JAK’s own tyrosine, leading to the dimerization of STAT3 (Heinrich et al., 2003). The second causes MAPK to become hyperphosphorylated as well as exhibit increased serine/threonine kinase activity via activating the Ras/Raf pathway (Bousoik and Montazeri Aliabadi, 2018). The third activates the PI3K-PKB/Akt pathway, enhancing NF-κB activity (Ait-Ghezala et al., 2007). Increased levels of IL-6 are linked to neurodegenerative diseases (Shan et al., 2024), and research studies reveal that PD patients’ blood and CSF have higher levels of IL-6 (Liu et al., 2022).

Studies have shown that IL-6 plays a key regulatory role in the neurotoxicity of PD (Kozina et al., 2022). Peripheral IL-6 levels significantly influence the neurotoxicity induced by LRRK2 mutations. For instance, in LRRK2-G2019S transgenic mice [where this mutation is a common pathogenic variant in familial Parkinson’s disease (Kozina et al., 2022; Omer et al., 2020), inhibiting IL-6 can alleviate the loss of nigral dopaminergic neurons induced by lipopolysaccharide (LPS) (Kozina et al., 2022)]. In the 6-hydroxydopamine (6-OHDA) model, overexpression of IL-6 exacerbates neuronal degeneration (Ma et al., 2020), suggesting a bidirectional interaction between LRRK2 dysfunction and the IL-6 signaling pathway. In the mechanism, IL-6 enhances the activity of acid-sensing ion channel 1a (ASIC1a) channels, allowing more calcium ions to enter neurons, leading to calcium overload and neuronal damage (Castellanos et al., 2024). Increased IL-6 levels have been observed in carriers of PRKN and PINK1 mutations, suggesting that both may act through a shared inflammatory pathway. This study provides the first evidence of elevated IL-6 in PINK1 mutation carriers, supporting its role in inflammation associated with mitochondrial autophagy (Borsche et al., 2020). Additionally, levels of IL-6 is correlated with a reduction in gray matter volume in the left precentral gyrus and the worsening of motor complication scores (UPDRS-IV) in PD patients (Chen et al., 2024), suggesting that PD motor symptoms may be aggravated by structural changes in specific brain regions. Research by Sterling et al. (2022) highlighted IL-6’s key role in mediating neuronal iron accumulation in α-syn-related PD. Experiments in α-syn mutant mice and in models with α-syn pre-formed fibrils (PFF) revealed that IL-6 is essential for iron accumulation through trans-IL-6 signaling. Moreover, microglia drive iron accumulation in this process by secreting IL-6 and other factors. Ultimately, IL-6 overexpression leads to iron accumulation (Sterling et al., 2022). IL-6 secreted by astrocytes in PD can induce neuronal death through conditioned media, an effect that can be inhibited by the IL-6R blocking antibody tocilizumab, indicating the crucial role of IL-6 in neuroinflammation and neurodegeneration in PD (Pons-Espinal et al., 2024). This highlights the pivotal role of IL-6 in neuroinflammatory neurodegeneration and potentially extends its significance to LRRK2-associated PD. An analysis also indicated that the risk of PD is considerably decreased by IL-6 inhibitors (P < 0.001) (Fu et al., 2024). These evidence supports the IL-6 signaling pathway as a potential therapeutic target for LRRK2-associated and other hereditary PD subtypes. It is evident that IL-6 plays a central role in LRRK2 mutation-mediated neurotoxicity and is intertwined with the pathways of hereditary and inflammatory PD. In summary, IL-6 plays an important role in the neuroinflammation of PD through various pathways (Figure 4). Targeting the IL-6 signaling pathway may offer neuroprotective strategies for different PD subtypes.

6 Conclusion and perspectives

6.1 Conclusion

Exercise exerts neuroprotective effects in PD by modulating multiple molecular factors. Myokines such as irisin and CTSB, secreted by skeletal muscles during physical activity, promote neurogenesis, mitochondrial function, and neuronal survival. Neurotrophic factors including BDNF, GDNF, and NT-3 support dopaminergic neuron maintenance and enhance synaptic plasticity. Furthermore, exercise downregulates pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α, alleviating neuroinflammation—a major contributor to PD progression. While it remains difficult to identify the most effective exercise type for regulating these factors, resistance training appears more effective in upregulating irisin, whereas aerobic exercise often enhances neurotrophic factor expression. Given the variability in individual response, exercise interventions should be tailored based on the patient’s physical status, preferences, and therapeutic goals to maximize benefits.

6.2 Limitations of existing research

Despite the widely recognized potential of exercise in treating PD, several limitations exist in current studies. First, detailed research on the specific molecular mechanisms of exercise, particularly how it mediates neuroprotective effects through factors like irisin, BDNF, and GDNF, is still lacking. Second, the effects of various forms of exercise on Parkinson’s patients are not entirely consistent and may even yield opposing effects in some cases. Moreover, individual differences such as disease progression, age, sex, and physical ability can significantly influence the effectiveness of exercise interventions. Most existing studies focus on specific populations, neglecting a comprehensive consideration of varying individual characteristics. Particularly for patients who have lost or have limited mobility, finding alternative therapeutic approaches (such as medications or physical therapy) that can simulate the neuroprotective effects of exercise remains a pressing issue. Furthermore, existing research predominantly addresses short-term exercise interventions, while not enough research has been done on the long-term consequences of exercising. Given that PD develops over time, symptoms and pathological changes worsen over time. Therefore, examining the long-term advantages of exercise and whether sustained exercise can continuously slow disease progression is a crucial avenue for further investigation.

6.3 The prospect of future research

Further exploration is needed regarding the relationship between cytokines and PD, particularly how various cytokines regulate neuronal plasticity and survival through specific signaling pathways. Additionally, investigating how to pharmacologically or otherwise simulate the effects of exercise-induced cytokines or enhance their activity within neurons may represent crucial strategies for patients unable to engage in exercise. Although the cytokines mentioned in this review are believed to be influenced by exercise and contribute positively to neuroprotection, it remains unclear whether they exhibit synergistic effects or act through different signaling pathways on dopaminergic neurons. Future research could further employ animal models or clinical trials to elucidate the mechanisms of these factors in PD patients and determine their possibility as targets for combined therapies. Furthermore, excessive exercise may induce oxidative stress, exacerbating neuroinflammation. Thus, understanding how to achieve a balance between anti-inflammatory and antioxidant systems during exercise will be an important focus. Specifically, research can investigate how different exercise intensities affect inflammatory cytokines to identify the optimal exercise dosage and minimize adverse reactions.

Individual differences among PD patients are significant; factors such as disease stage, physical constitution, and exercise ability may all influence treatment efficacy. Therefore, future studies should delve deeper into personalized exercise intervention programs to ensure that diverse patient groups might benefit from exercise. As the disease progresses, the severity of symptoms varies. For early-stage patients, exercise can serve as a preventive intervention to delay disease progression, whereas tailored exercise programs are needed for mid- to late-stage patients to address motor symptoms and alleviate non-motor symptoms. For example, mild patients may engage in moderate to high-intensity aerobic exercises to maximize neurotrophic factor secretion, while moderate to severe patients may benefit more from low-intensity rehabilitative training to avoid overburdening their bodies. Given the significant variability in exercise capacity among patients, establishing an individualized assessment system for exercise ability is essential to determining the appropriate intensity for each patient. Future research could develop assessment tools to evaluate various parameters such as cardiopulmonary function, muscle strength, and balance, thereby facilitating the creation of personalized exercise prescriptions. Different forms of exercise can specifically target various Parkinson’s symptoms. For instance, yoga and Tai Chi can enhance postural stability and flexibility; treadmill training can improve gait and coordination; and inspiratory muscle training can help alleviate dysphagia and other non-motor symptoms. Future studies can design targeted exercise programs based on the specific symptoms presented by patients to maximize the efficacy of exercise interventions.

In short, exercise has demonstrated promising effects in reducing the risk of developing PD and in alleviating its symptoms. However, the underlying molecular mechanisms through which exercise confers neuroprotective effects remain incompletely understood. Uncovering these mechanisms will require sustained, in-depth research efforts. A better understanding of these pathways may help integrate exercise more effectively into comprehensive treatment strategies, ultimately enabling more individuals with PD to benefit from its therapeutic potential.

Author contributions

W-QL: Writing – original draft, Writing – review & editing. J-HY: Writing – original draft. L-LL: Writing – original draft. S-TD: Project administration, Writing – review & editing. BY: Validation, Writing – review & editing. LJ: Writing – review & editing. NY: Writing – review & editing. H-DL: Writing – review & editing, Funding acquisition, Project administration, Resources, Supervision.

Funding

The authors declare that financial support was received for the research and/or publication of this article. This research was supported by the Natural Science Foundation Project of Chongqing Yuzhong District, grant No. 20240130, the Project of Youth Innovative Talents Training Program of Chongqing under grant No. CY220404, and the Tutorial System of Medical Undergraduate Foundation of Experimental Teaching Center in Chongqing Medical University under grant Nos. LTMCMTS202116 and LTMCMTS202202.

Acknowledgments

The figures were created using BioRender.com.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abrous, D. N., and Wojtowicz, J. M. (2015). Interaction between neurogenesis and hippocampal memory system: New vistas. Cold Spring Harb. Perspect. Biol. 7:a018952. doi: 10.1101/cshperspect.a018952

PubMed Abstract | Crossref Full Text | Google Scholar

Ahmad, M. H., Rizvi, M. A., Ali, M., and Mondal, A. C. (2023). Neurobiology of depression in Parkinson’s disease: Insights into epidemiology, molecular mechanisms and treatment strategies. Ageing Res. Rev. 85:101840. doi: 10.1016/j.arr.2022.101840

PubMed Abstract | Crossref Full Text | Google Scholar

Ait-Ghezala, G., Volmar, C. H., Frieling, J., Paris, D., Tweed, M., Bakshi, P., et al. (2007). CD40 promotion of amyloid beta production occurs via the NF-kappaB pathway. Eur. J. Neurosci. 25, 1685–1695. doi: 10.1111/j.1460-9568.2007.05424.x

PubMed Abstract | Crossref Full Text | Google Scholar

Albrecht, E., Norheim, F., Thiede, B., Holen, T., Ohashi, T., Schering, L., et al. (2015). Irisin - a myth rather than an exercise-inducible myokine. Sci. Rep. 5:8889. doi: 10.1038/srep08889

PubMed Abstract | Crossref Full Text | Google Scholar

Algul, S., Ozdenk, C., and Ozcelik, O. (2017). Variations in leptin, nesfatin-1 and irisin levels induced by aerobic exercise in young trained and untrained male subjects. Biol. Sport 34, 339–344. doi: 10.5114/biolsport.2017.69821

PubMed Abstract | Crossref Full Text | Google Scholar

Ali, N. H., Alhamdan, N. A., Al-Kuraishy, H. M., Al-Gareeb, A. I., Elekhnawy, E., and Batiha, G. E. (2024). Irisin/PGC-1α/FNDC5 pathway in Parkinson’s disease: Truth under the throes. Naunyn Schmiedebergs Arch. Pharmacol. 397, 1985–1995. doi: 10.1007/s00210-023-02726-9

PubMed Abstract | Crossref Full Text | Google Scholar

Allen, S. J., Watson, J. J., Shoemark, D. K., Barua, N. U., and Patel, N. K. (2013). GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol. Ther. 138, 155–175. doi: 10.1016/j.pharmthera.2013.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

Allwright, M., Mundell, H., Sutherland, G., Austin, P., and Guennewig, B. (2023). Machine learning analysis of the UK Biobank reveals IGF-1 and inflammatory biomarkers predict Parkinson’s disease risk. PLoS One 18:e0285416. doi: 10.1371/journal.pone.0285416

PubMed Abstract | Crossref Full Text | Google Scholar

Amin, R., Quispe, C., Docea, A. O., Ydyrys, A., Kulbayeva, M., Durna Daştan, S., et al. (2022). The role of Tumour Necrosis Factor in neuroinflammation associated with Parkinson’s disease and targeted therapies. Neurochem. Int. 158:105376. doi: 10.1016/j.neuint.2022.105376

PubMed Abstract | Crossref Full Text | Google Scholar

An, X. K., Peng, R., Li, T., Burgunder, J. M., Wu, Y., Chen, W. J., et al. (2008). LRRK2 Gly2385Arg variant is a risk factor of Parkinson’s disease among Han-Chinese from mainland China. Eur. J. Neurol. 15, 301–305. doi: 10.1111/j.1468-1331.2007.02052.x

PubMed Abstract | Crossref Full Text | Google Scholar

Andreska, T., Aufmkolk, S., Sauer, M., and Blum, R. (2014). High abundance of BDNF within glutamatergic presynapses of cultured hippocampal neurons. Front. Cell Neurosci. 8:107. doi: 10.3389/fncel.2014.00107

PubMed Abstract | Crossref Full Text | Google Scholar

Armstrong, M. J., and Okun, M. S. (2020). Diagnosis and treatment of parkinson disease: A review. JAMA 323, 548–560. doi: 10.1001/jama.2019.22360

PubMed Abstract | Crossref Full Text | Google Scholar

Bae, E. J., Choi, M., Kim, J. T., Kim, D. K., Jung, M. K., Kim, C., et al. (2022). TNF-α promotes α-synuclein propagation through stimulation of senescence-associated lysosomal exocytosis. Exp. Mol. Med. 54, 788–800. doi: 10.1038/s12276-022-00789-x

PubMed Abstract | Crossref Full Text | Google Scholar

Baghi, M., Yadegari, E., Rostamian Delavar, M., Peymani, M., Ganjalikhani-Hakemi, M., Salari, M., et al. (2021). MiR-193b deregulation is associated with Parkinson’s disease. J. Cell Mol. Med. 25, 6348–6360. doi: 10.1111/jcmm.16612

PubMed Abstract | Crossref Full Text | Google Scholar

Bahmani, E., Hoseini, R., and Amiri, E. (2022). Home-based aerobic training and vitamin D improve neurotrophins and inflammatory biomarkers in MS patients. Mult. Scler. Relat. Disord. 60:103693. doi: 10.1016/j.msard.2022.103693

PubMed Abstract | Crossref Full Text | Google Scholar

Bai, L., Tan, C., Ren, J., Liu, J., Zou, W., Liu, G., et al. (2023). Cordyceps militaris acidic polysaccharides improve learning and memory impairment in mice with exercise fatigue through the PI3K/NRF2/HO-1 signalling pathway. Int. J. Biol. Macromol. 227, 158–172. doi: 10.1016/j.ijbiomac.2022.12.071

PubMed Abstract | Crossref Full Text | Google Scholar

Barcia, C., Ros, C. M., Annese, V., Gómez, A., Ros-Bernal, F., Aguado-Yera, D., et al. (2011). IFN-γ signaling, with the synergistic contribution of TNF-α, mediates cell specific microglial and astroglial activation in experimental models of Parkinson’s disease. Cell Death Dis. 2:e142. doi: 10.1038/cddis.2011.17

PubMed Abstract | Crossref Full Text | Google Scholar

Barker, R. A., Björklund, A., Gash, D. M., Whone, A., Van Laar, A., Kordower, J. H., et al. (2020). GDNF and Parkinson’s disease: Where next? a summary from a recent workshop. J. Park. Dis. 10, 875–891. doi: 10.3233/JPD-202004

PubMed Abstract | Crossref Full Text | Google Scholar

Bastioli, G., Arnold, J. C., Mancini, M., Mar, A. C., Gamallo-Lana, B., Saadipour, K., et al. (2022). Voluntary exercise boosts striatal dopamine release: Evidence for the necessary and sufficient role of BDNF. J. Neurosci. 42, 4725–4736. doi: 10.1523/JNEUROSCI.2273-21.2022

PubMed Abstract | Crossref Full Text | Google Scholar

Bazzari, A. H., and Bazzari, F. H. (2022). BDNF therapeutic mechanisms in neuropsychiatric disorders. Int. J. Mol. Sci. 23:8417. doi: 10.3390/ijms23158417

PubMed Abstract | Crossref Full Text | Google Scholar

Bednarski, E., Lauterborn, J. C., Gall, C. M., and Lynch, G. (1998). Lysosomal dysfunction reduces brain-derived neurotrophic factor expression. Exp. Neurol. 150, 128–135. doi: 10.1006/exnr.1997.6747

PubMed Abstract | Crossref Full Text | Google Scholar

Behrendt, T., Kirschnick, F., Kröger, L., Beileke, P., Rezepin, M., Brigadski, T., et al. (2021). Comparison of the effects of open vs. closed skill exercise on the acute and chronic BDNF, IGF-1 and IL-6 response in older healthy adults. BMC Neurosci. 22:71. doi: 10.1186/s12868-021-00675-8

PubMed Abstract | Crossref Full Text | Google Scholar

Bek, J., Arakaki, A. I., Lawrence, A., Sullivan, M., Ganapathy, G., and Poliakoff, E. (2020). Dance and Parkinson’s: A review and exploration of the role of cognitive representations of action. Neurosci. Biobehav. Rev. 109, 16–28. doi: 10.1016/j.neubiorev.2019.12.023

PubMed Abstract | Crossref Full Text | Google Scholar

Bellomo, G., Paciotti, S., Gatticchi, L., and Parnetti, L. (2020). The vicious cycle between α-synuclein aggregation and autophagic-lysosomal dysfunction. Mov. Disord. 35, 34–44. doi: 10.1002/mds.27895

PubMed Abstract | Crossref Full Text | Google Scholar

Benedini, S., Dozio, E., Invernizzi, P. L., Vianello, E., Banfi, G., Terruzzi, I., et al. (2017). Irisin: A potential link between physical exercise and metabolism-an observational study in differently trained subjects, from elite athletes to sedentary people. J. Diab. Res. 2017:1039161. doi: 10.1155/2017/1039161

PubMed Abstract | Crossref Full Text | Google Scholar

Ben-Shlomo, Y., Darweesh, S., Llibre-Guerra, J., Marras, C., San Luciano, M., and Tanner, C. (2024). The epidemiology of Parkinson’s disease. Lancet 403, 283–292. doi: 10.1016/S0140-6736(23)01419-8

PubMed Abstract | Crossref Full Text | Google Scholar

Bhadra, J., Sridhar, N., Fajrial, A. K., Hammond, N., Xue, D., and Ding, X. (2023). Acoustic streaming enabled moderate swimming exercise reduces neurodegeneration in C. elegans. Sci. Adv. 9:eadf5056. doi: 10.1126/sciadv.adf5056

PubMed Abstract | Crossref Full Text | Google Scholar

Bizjak, D. A., Zügel, M., Treff, G., Winkert, K., Jerg, A., Hudemann, J., et al. (2021). Effects of training status and exercise mode on global gene expression in skeletal muscle. Int. J. Mol. Sci. 22:12578. doi: 10.3390/ijms222212578

PubMed Abstract | Crossref Full Text | Google Scholar

Björklund, A. (2021). GDNF therapy: Can we make it work? J. Park. Dis. 11, 1019–1022. doi: 10.3233/JPD-212706

PubMed Abstract | Crossref Full Text | Google Scholar

Blum, A., and Miller, H. (1998). Role of cytokines in heart failure. Am. Heart J. 135(2 Pt 1), 181–186. doi: 10.1016/s0002-8703(98)70080-8

PubMed Abstract | Crossref Full Text | Google Scholar

Bognar, S., DeFaria, A. M., O’Dwyer, C., Pankiw, E., Simic Bogler, J., Teixeira, S., et al. (2017). More than just dancing: Experiences of people with Parkinson’s disease in a therapeutic dance program. Disabil. Rehabil. 39, 1073–1078. doi: 10.1080/09638288.2016.1175037

PubMed Abstract | Crossref Full Text | Google Scholar

Bonafina, A., Trinchero, M. F., Ríos, A. S., Bekinschtein, P., Schinder, A. F., Paratcha, G., et al. (2019). GDNF and GFRα1 are required for proper integration of adult-born hippocampal neurons. Cell Rep. 29, 4308–4319.e4. doi: 10.1016/j.celrep.2019.11.100.

PubMed Abstract | Crossref Full Text | Google Scholar

Borsche, M., König, I. R., Delcambre, S., Petrucci, S., Balck, A., Brüggemann, N., et al. (2020). Mitochondrial damage-associated inflammation highlights biomarkers in PRKN/PINK1 parkinsonism. Brain 143, 3041–3051. doi: 10.1093/brain/awaa246

PubMed Abstract | Crossref Full Text | Google Scholar

Boström, P., Wu, J., Jedrychowski, M. P., Korde, A., Ye, L., Lo, J. C., et al. (2012). A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463–468. doi: 10.1038/nature10777

PubMed Abstract | Crossref Full Text | Google Scholar

Bousoik, E., and Montazeri Aliabadi, H. (2018). “Do we know jack” about JAK? a closer look at JAK/STAT signaling pathway. Front. Oncol. 8:287. doi: 10.3389/fonc.2018.00287

PubMed Abstract | Crossref Full Text | Google Scholar

Bras, J., Guerreiro, R., Darwent, L., Parkkinen, L., Ansorge, O., Escott-Price, V., et al. (2014). Genetic analysis implicates APOE, SNCA and suggests lysosomal dysfunction in the etiology of dementia with Lewy bodies. Hum. Mol. Genet. 23, 6139–6146. doi: 10.1093/hmg/ddu334

PubMed Abstract | Crossref Full Text | Google Scholar

Brenmoehl, J., Albrecht, E., Komolka, K., Schering, L., Langhammer, M., Hoeflich, A., et al. (2014). Irisin is elevated in skeletal muscle and serum of mice immediately after acute exercise. Int. J. Biol. Sci. 10, 338–349. doi: 10.7150/ijbs.7972

PubMed Abstract | Crossref Full Text | Google Scholar

Bruno, M. A., and Cuello, A. C. (2006). Activity-dependent release of precursor nerve growth factor, conversion to mature nerve growth factor, and its degradation by a protease cascade. Proc. Natl. Acad. Sci. U. S. A. 103, 6735–6740. doi: 10.1073/pnas.0510645103

PubMed Abstract | Crossref Full Text | Google Scholar

Buehlmeyer, K., Doering, F., Daniel, H., Petridou, A., Mougios, V., Schulz, T., et al. (2007). IGF-1 gene expression in rat colonic mucosa after different exercise volumes. J. Sports Sci. Med. 6, 434–440.

Google Scholar

Cabezas, R., Baez-Jurado, E., Hidalgo-Lanussa, O., Echeverria, V., Ashrad, G. M., Sahebkar, A., et al. (2019). Growth factors and neuroglobin in astrocyte protection against neurodegeneration and oxidative stress. Mol. Neurobiol. 56, 2339–2351. doi: 10.1007/s12035-018-1203-9

PubMed Abstract | Crossref Full Text | Google Scholar

Calissano, P., and Levi-Montalcini, R. (1979). Is NGF an enzyme? Nature 280:359. doi: 10.1038/280359a0

PubMed Abstract | Crossref Full Text | Google Scholar

Calvo, P. M., Hernández, R. G., de la Cruz, R. R., and Pastor, A. M. (2022). VEGF is an essential retrograde trophic factor for motoneurons. Proc. Natl. Acad. Sci. U. S. A. 119:e2202912119. doi: 10.1073/pnas.2202912119

PubMed Abstract | Crossref Full Text | Google Scholar

Calvo, P. M., Hernández, R. G., Pastor, A. M., and de la Cruz, , R. R. (2024). VEGF and neuronal survival. Neuroscientist 30, 71–86. doi: 10.1177/10738584221120803

PubMed Abstract | Crossref Full Text | Google Scholar

Carswell, E. A., Old, L. J., Kassel, R. L., Green, S., Fiore, N., and Williamson, B. (1975). An endotoxin-induced serum factor that causes necrosis of tumors. Proc. Natl. Acad. Sci. U. S. A. 72, 3666–3670. doi: 10.1073/pnas.72.9.3666

PubMed Abstract | Crossref Full Text | Google Scholar

Cassilhas, R. C., Lee, K. S., Fernandes, J., Oliveira, M. G., Tufik, S., Meeusen, R., et al. (2012). Spatial memory is improved by aerobic and resistance exercise through divergent molecular mechanisms. Neuroscience 202, 309–317. doi: 10.1016/j.neuroscience.2011.11.029

PubMed Abstract | Crossref Full Text | Google Scholar

Castellanos, L. C. S., Gatto, R. G., Malnati, G. O. M., Montes, M. M., Uchitel, O. D., and Weissmann, C. (2024). Redistribution of ASIC1a channels triggered by IL-6: Potential role of ASIC1a in neuroinflammation. Biochim Biophys. Acta Mol. Basis Dis. 1870:166927. doi: 10.1016/j.bbadis.2023.166927

PubMed Abstract | Crossref Full Text | Google Scholar

Chao, M. V., Rajagopal, R., and Lee, F. S. (2006). Neurotrophin signalling in health and disease. Clin. Sci. 110, 167–173. doi: 10.1042/CS20050163

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, B., Zhou, H., Liu, X., Yang, W., Luo, Y., Zhu, S., et al. (2024). Correlations of gray matter volume with peripheral cytokines in Parkinson’s disease. Neurobiol. Dis. 201:106693. doi: 10.1016/j.nbd.2024.106693

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, C., Zhou, M., Ge, Y., and Wang, X. (2020). SIRT1 and aging related signaling pathways. Mech. Ageing Dev. 187:111215. doi: 10.1016/j.mad.2020.111215

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, H. T., Chung, Y. C., Chen, Y. J., Ho, S. Y., and Wu, H. J. (2017). Effects of different types of exercise on body composition, muscle strength, and IGF-1 in the elderly with sarcopenic obesity. J. Am. Geriatr. Soc. 65, 827–832. doi: 10.1111/jgs.14722

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, W., Hu, Y., and Ju, D. (2020). Gene therapy for neurodegenerative disorders: Advances, insights and prospects. Acta Pharm. Sin. B 10, 1347–1359. doi: 10.1016/j.apsb.2020.01.015

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, X., Chen, A., Wei, J., Huang, Y., Deng, J., Chen, P., et al. (2023). Dexmedetomidine alleviates cognitive impairment by promoting hippocampal neurogenesis via BDNF/TrkB/CREB signaling pathway in hypoxic-ischemic neonatal rats. CNS Neurosci. Ther. 30:e14486. doi: 10.1111/cns.14486

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, X., Hu, Y., Cao, Z., Liu, Q., and Cheng, Y. (2018). Cerebrospinal fluid inflammatory cytokine aberrations in Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis: A systematic review and meta-analysis. Front. Immunol. 9:2122. doi: 10.3389/fimmu.2018.02122

PubMed Abstract | Crossref Full Text | Google Scholar

Chmielarz, P., Er, Ş,Konovalova, J., Bandres, L., Hlushchuk, I., Albert, K., et al. (2020). GDNF/RET signaling pathway activation eliminates lewy body pathology in midbrain dopamine neurons. Mov. Disord. 35, 2279–2289. doi: 10.1002/mds.28258

PubMed Abstract | Crossref Full Text | Google Scholar

Church, D. D., Hoffman, J. R., Mangine, G. T., Jajtner, A. R., Townsend, J. R., Beyer, K. S., et al. (2016). Comparison of high-intensity vs. high-volume resistance training on the BDNF response to exercise. J. Appl. Physiol. 121, 123–128. doi: 10.1152/japplphysiol.00233.2016

PubMed Abstract | Crossref Full Text | Google Scholar

Ciafrè, S., Ferraguti, G., Tirassa, P., Iannitelli, A., Ralli, M., Greco, A., et al. (2020). Nerve growth factor in the psychiatric brain. Riv. Psichiatr. 55, 4–15. doi: 10.1708/3301.32713

PubMed Abstract | Crossref Full Text | Google Scholar

Clarke, L. E., Liddelow, S. A., Chakraborty, C., Münch, A. E., Heiman, M., and Barres, B. A. (2018). Normal aging induces A1-like astrocyte reactivity. Proc. Natl. Acad. Sci. U. S. A. 115, E1896–E1905. doi: 10.1073/pnas.1800165115

PubMed Abstract | Crossref Full Text | Google Scholar

Claus, I., Muhle, P., Czechowski, J., Ahring, S., Labeit, B., Suntrup-Krueger, S., et al. (2021). Expiratory muscle strength training for therapy of pharyngeal dysphagia in Parkinson’s disease. Mov. Disord. 36, 1815–1824. doi: 10.1002/mds.28552

PubMed Abstract | Crossref Full Text | Google Scholar

Colucci-D’Amato, L., Speranza, L., and Volpicelli, F. (2020). Neurotrophic factor BDNF, physiological functions and therapeutic potential in depression, neurodegeneration and brain Cancer. Int. J. Mol. Sci. 21:7777. doi: 10.3390/ijms21207777

PubMed Abstract | Crossref Full Text | Google Scholar

Cook, D. A., Kannarkat, G. T., Cintron, A. F., Butkovich, L. M., Fraser, K. B., Chang, J., et al. (2017). LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis. 3:11. doi: 10.1038/s41531-017-0010-8

PubMed Abstract | Crossref Full Text | Google Scholar

Cosio, P. L., Moreno-Simonet, L., Porcelli, A., Lloret, M., Padulles, X., Padulles, J. M., et al. (2024). Assessment of inter-individual variability in hamstring muscle recovery after a sport-specific sprint training in women and men. Front. Physiol. 14:1331878. doi: 10.3389/fphys.2023.1331878

PubMed Abstract | Crossref Full Text | Google Scholar

Côté, M. P., Azzam, G. A., Lemay, M. A., Zhukareva, V., and Houlé, J. D. (2011). Activity-dependent increase in neurotrophic factors is associated with an enhanced modulation of spinal reflexes after spinal cord injury. J. Neurotrauma 28, 299–309. doi: 10.1089/neu.2010.1594

PubMed Abstract | Crossref Full Text | Google Scholar

Daviaud, N., Garbayo, E., Sindji, L., Martínez-Serrano, A., Schiller, P. C., and Montero-Menei, C. N. (2015). Survival, differentiation, and neuroprotective mechanisms of human stem cells complexed with neurotrophin-3-releasing pharmacologically active microcarriers in an ex vivo model of Parkinson’s disease. Stem Cells Transl. Med. 4, 670–684. doi: 10.5966/sctm.2014-0139

PubMed Abstract | Crossref Full Text | Google Scholar

Davis, A. A., Inman, C. E., Wargel, Z. M., Dube, U., Freeberg, B. M., Galluppi, A., et al. (2020). APOE genotype regulates pathology and disease progression in synucleinopathy. Sci. Transl. Med. 12:eaay3069. doi: 10.1126/scitranslmed.aay3069

PubMed Abstract | Crossref Full Text | Google Scholar

de Azambuja, G., Hortscht, U., Hoheisel, U., Oliveira Fusaro, M. C., Mense, S., and Treede, R. D. (2018). Short-term swimming exercise attenuates the sensitization of dorsal horn neurons in rats with NGF-induced low back pain. Eur. J. Pain 22, 1409–1418. doi: 10.1002/ejp.1230

PubMed Abstract | Crossref Full Text | Google Scholar

De Sousa, R. A. L. (2024). Exercise-produced irisin effects on brain-related pathological conditions. Metab. Brain Dis. 39, 1679–1687. doi: 10.1007/s11011-024-01412-w

PubMed Abstract | Crossref Full Text | Google Scholar

D’Ercole, A. J., Ye, P., Calikoglu, A. S., and Gutierrez-Ospina, G. (1996). The role of the insulin-like growth factors in the central nervous system. Mol. Neurobiol. 13, 227–255. doi: 10.1007/BF02740625

PubMed Abstract | Crossref Full Text | Google Scholar

Devi, S. A., and Kiran, T. R. (2004). Regional responses in antioxidant system to exercise training and dietary vitamin E in aging rat brain. Neurobiol. Aging 25, 501–508. doi: 10.1016/S0197-4580(03)00112-X

PubMed Abstract | Crossref Full Text | Google Scholar

Dinoff, A., Herrmann, N., Swardfager, W., and Lanctôt, K. L. (2017). The effect of acute exercise on blood concentrations of brain-derived neurotrophic factor in healthy adults: A meta-analysis. Eur. J. Neurosci. 46, 1635–1646. doi: 10.1111/ejn.13603

PubMed Abstract | Crossref Full Text | Google Scholar

Dinoff, A., Herrmann, N., Swardfager, W., Liu, C. S., Sherman, C., Chan, S., et al. (2016). The effect of exercise training on resting concentrations of peripheral Brain-Derived Neurotrophic Factor (BDNF): A meta-analysis. PLoS One 11:e0163037. doi: 10.1371/journal.pone.0163037

PubMed Abstract | Crossref Full Text | Google Scholar

Dorsey, E. R., Sherer, T., Okun, M. S., and Bloem, B. R. (2018). The emerging evidence of the Parkinson pandemic. J. Parkinsons Dis. 8, S3–S8. doi: 10.3233/JPD-181474

PubMed Abstract | Crossref Full Text | Google Scholar

Drobny, A., Boros, F. A., Balta, D., Prieto Huarcaya, S., Caylioglu, D., Qazi, N., et al. (2023). Reciprocal effects of alpha-synuclein aggregation and lysosomal homeostasis in synucleinopathy models. Transl. Neurodegener. 12:31. doi: 10.1186/s40035-023-00363-z

PubMed Abstract | Crossref Full Text | Google Scholar

Dunleavy, C., Elsworthy, R. J., Wood, S. J., Allott, K., Spencer, F., Upthegrove, R., et al. (2024). Exercise4Psychosis: A randomised control trial assessing the effect of moderate-to-vigorous exercise on inflammatory biomarkers and negative symptom profiles in men with first-episode psychosis. Brain Behav. Immun. 120, 379–390. doi: 10.1016/j.bbi.2024.06.017

PubMed Abstract | Crossref Full Text | Google Scholar

Dupont-Versteegden, E. E., Houlé, J. D., Dennis, R. A., Zhang, J., Knox, M., Wagoner, G., et al. (2004). Exercise-induced gene expression in soleus muscle is dependent on time after spinal cord injury in rats. Muscle Nerve 29, 73–81. doi: 10.1002/mus.10511

PubMed Abstract | Crossref Full Text | Google Scholar

Dupuis, O., Girardie, J., Van Gaever, M., Garnier, P., Coq, J. O., Canu, M. H., et al. (2024). Early movement restriction affects FNDC5/Irisin and BDNF levels in rat muscle and brain. Int. J. Mol. Sci. 25:3918. doi: 10.3390/ijms25073918

PubMed Abstract | Crossref Full Text | Google Scholar

Dutta, D., Paidi, R. K., Raha, S., Roy, A., Chandra, S., and Pahan, K. (2022). Treadmill exercise reduces α-synuclein spreading via PPARα. Cell Rep. 40:111058. doi: 10.1016/j.celrep.2022.111058

PubMed Abstract | Crossref Full Text | Google Scholar

Ebert, A. D., Beres, A. J., Barber, A. E., and Svendsen, C. N. (2008). Human neural progenitor cells over-expressing IGF-1 protect dopamine neurons and restore function in a rat model of Parkinson’s disease. Exp. Neurol. 209, 213–223. doi: 10.1016/j.expneurol.2007.09.022

PubMed Abstract | Crossref Full Text | Google Scholar

Eguchi, T., Kuwahara, T., Sakurai, M., Komori, T., Fujimoto, T., Ito, G., et al. (2018). LRRK2 and its substrate Rab GTPases are sequentially targeted onto stressed lysosomes and maintain their homeostasis. Proc. Natl. Acad. Sci. U. S. A. 115, E9115–E9124. doi: 10.1073/pnas.1812196115

PubMed Abstract | Crossref Full Text | Google Scholar

El Hayek, L., Khalifeh, M., Zibara, V., Abi Assaad, R., Emmanuel, N., Karnib, N., et al. (2019). Lactate mediates the effects of exercise on learning and memory through SIRT1-Dependent activation of hippocampal Brain-Derived Neurotrophic Factor (BDNF). J. Neurosci. 39, 2369–2382. doi: 10.1523/JNEUROSCI.1661-18.2019

PubMed Abstract | Crossref Full Text | Google Scholar

Eldeeb, M. A., Thomas, R. A., Ragheb, M. A., Fallahi, A., and Fon, E. A. (2022). Mitochondrial quality control in health and in Parkinson’s disease. Physiol. Rev. 102, 1721–1755. doi: 10.1152/physrev.00041.2021

PubMed Abstract | Crossref Full Text | Google Scholar

ElGrawani, W., Sun, G., Kliem, F. P., Sennhauser, S., Pierre-Ferrer, S., Rosi-Andersen, A., et al. (2024). BDNF-TrkB signaling orchestrates the buildup process of local sleep. Cell Rep. 43:114500. doi: 10.1016/j.celrep.2024.114500

PubMed Abstract | Crossref Full Text | Google Scholar

Fabel, K., Wolf, S. A., Ehninger, D., Babu, H., Leal-Galicia, P., and Kempermann, G. (2009). Additive effects of physical exercise and environmental enrichment on adult hippocampal neurogenesis in mice. Front. Neurosci. 3:50. doi: 10.3389/neuro.22.002.2009

PubMed Abstract | Crossref Full Text | Google Scholar

Fallah Mohammadi, Z., Falah Mohammadi, H., and Patel, D. I. (2019). Comparing the effects of progressive and mild intensity treadmill running protocols on neuroprotection of parkinsonian rats. Life Sci. 229, 219–224. doi: 10.1016/j.lfs.2019.05.036

PubMed Abstract | Crossref Full Text | Google Scholar

Feng, L., Li, B., Xi, Y., Cai, M., and Tian, Z. (2022). Aerobic exercise and resistance exercise alleviate skeletal muscle atrophy through IGF-1/IGF-1R-PI3K/Akt pathway in mice with myocardial infarction. Am. J. Physiol. Cell Physiol. 322, C164–C176. doi: 10.1152/ajpcell.00344.2021

PubMed Abstract | Crossref Full Text | Google Scholar

Finan, J. D., Udani, S. V., Patel, V., and Bailes, J. E. (2018). The influence of the Val66Met polymorphism of brain-derived neurotrophic factor on neurological function after traumatic brain injury. J. Alzheimers Dis. 65, 1055–1064. doi: 10.3233/JAD-180585

PubMed Abstract | Crossref Full Text | Google Scholar

Fiorenza, M., Gliemann, L., Brandt, N., and Bangsbo, J. (2020). Hormetic modulation of angiogenic factors by exercise-induced mechanical and metabolic stress in human skeletal muscle. Am. J. Physiol. Heart Circ. Physiol. 319, H824–H834. doi: 10.1152/ajpheart.00432.2020

PubMed Abstract | Crossref Full Text | Google Scholar

Folkerts, A. K., Nielsen, J., Gollan, R., Lansu, A., Solfronk, D., Monsef, I., et al. (2023). Physical exercise as a potential treatment for fatigue in parkinson’s disease? a systematic review and meta-analysis of pharmacological and non-pharmacological interventions. J. Parkinsons Dis. 13, 659–679. doi: 10.3233/JPD-225116

PubMed Abstract | Crossref Full Text | Google Scholar

Fu, C., Wang, L., and Cai, W. (2024). IL6 receptor inhibitors: Exploring the therapeutic potential across multiple diseases through drug target Mendelian randomization. Front. Immunol. 15:1452849. doi: 10.3389/fimmu.2024.1452849

PubMed Abstract | Crossref Full Text | Google Scholar

Gaenslen, A., Swid, I., Liepelt-Scarfone, I., Godau, J., and Berg, D. (2011). The patients’ perception of prodromal symptoms before the initial diagnosis of Parkinson’s disease. Mov. Disord. 26, 653–658. doi: 10.1002/mds.23499

PubMed Abstract | Crossref Full Text | Google Scholar

Geng, X., Zou, Y., Li, J., Li, S., Qi, R., Yu, H., et al. (2023). BDNF alleviates Parkinson’s disease by promoting STAT3 phosphorylation and regulating neuronal autophagy. Cell Tissue Res. 393, 455–470. doi: 10.1007/s00441-023-03806-1

PubMed Abstract | Crossref Full Text | Google Scholar

Georgievska, B., Kirik, D., and Björklund, A. (2004). Overexpression of glial cell line-derived neurotrophic factor using a lentiviral vector induces time- and dose-dependent downregulation of tyrosine hydroxylase in the intact nigrostriatal dopamine system. J. Neurosci. 24, 6437–6445. doi: 10.1523/JNEUROSCI.1122-04.2004

PubMed Abstract | Crossref Full Text | Google Scholar

Godau, J., Herfurth, M., Kattner, B., Gasser, T., and Berg, D. (2010). Increased serum insulin-like growth factor 1 in early idiopathic Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 81, 536–538. doi: 10.1136/jnnp.2009.175752

PubMed Abstract | Crossref Full Text | Google Scholar

Guazzarini, A. G., Mancinetti, F., Bastiani, P., Scamosci, M., Cecchetti, R., Boccardi, V., et al. (2024). Tai chi, irisin and cognitive performance: A clinical and biological investigation in older adults. Aging Clin. Exp. Res. 36:90. doi: 10.1007/s40520-024-02743-5

PubMed Abstract | Crossref Full Text | Google Scholar

Guo, H., Xia, D., Liao, S., Niu, B., Tang, J., Hu, H., et al. (2019). Vascular endothelial growth factor improves the cognitive decline of Alzheimer’s disease via concurrently inducing the expression of ADAM10 and reducing the expression of β-site APP cleaving enzyme 1 in Tg2576 mice. Neurosci. Res. 142, 49–57. doi: 10.1016/j.neures.2018.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

Guo, H., Zhou, H., Lu, J., Qu, Y., Yu, D., and Tong, Y. (2016). Vascular endothelial growth factor: An attractive target in the treatment of hypoxic/ischemic brain injury. Neural Regen. Res. 11, 174–179. doi: 10.4103/1673-5374.175067

PubMed Abstract | Crossref Full Text | Google Scholar

Gupta, V., Chitranshi, N., and Gupta, V. B. (2024). Genetic risk, inflammation, and therapeutics: An editorial overview of recent advances in aging brains and neurodegeneration. Aging Dis. 15, 1989–1993. doi: 10.14336/AD.2024.0986

PubMed Abstract | Crossref Full Text | Google Scholar

Guzmán-Sastoque, P., Sotelo, S., Esmeral, N. P., Albarracín, S. L., Sutachan, J. J., Reyes, L. H., et al. (2024). Assessment of CRISPRa-mediated gdnf overexpression in an In vitro Parkinson’s disease model. Front. Bioeng. Biotechnol. 12:1420183. doi: 10.3389/fbioe.2024.1420183

PubMed Abstract | Crossref Full Text | Google Scholar

Gyorkos, A. M., McCullough, M. J., and Spitsbergen, J. M. (2014). Glial cell line-derived neurotrophic factor (GDNF) expression and NMJ plasticity in skeletal muscle following endurance exercise. Neuroscience 257, 111–118. doi: 10.1016/j.neuroscience.2013.10.068

PubMed Abstract | Crossref Full Text | Google Scholar

Harro, C. C., Shoemaker, M. J., Coatney, C. M., Lentine, V. E., Lieffers, L. R., Quigley, J. J., et al. (2022). Effects of nordic walking exercise on gait, motor/non-motor symptoms, and serum brain-derived neurotrophic factor in individuals with Parkinson’s disease. Front. Rehabil. Sci. 3:1010097. doi: 10.3389/fresc.2022.1010097

PubMed Abstract | Crossref Full Text | Google Scholar

Hecking, I., Stegemann, L. N., Theis, V., Vorgerd, M., Matschke, V., Stahlke, S., et al. (2022). Neuroprotective effects of VEGF in the enteric nervous system. Int. J. Mol. Sci. 23:6756. doi: 10.3390/ijms23126756

PubMed Abstract | Crossref Full Text | Google Scholar

Heinrich, P. C., Behrmann, I., Haan, S., Hermanns, H. M., Müller-Newen, G., and Schaper, F. (2003). Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem. J. 374(Pt 1), 1–20. doi: 10.1042/BJ20030407

PubMed Abstract | Crossref Full Text | Google Scholar

Hernandez-Chan, N. G., Bannon, M. J., Orozco-Barrios, C. E., Escobedo, L., Zamudio, S., De la Cruz, F., et al. (2015). Neurotensin-polyplex-mediated brain-derived neurotrophic factor gene delivery into nigral dopamine neurons prevents nigrostriatal degeneration in a rat model of early Parkinson’s disease. J. Biomed. Sci. 22:59. doi: 10.1186/s12929-015-0166-7

PubMed Abstract | Crossref Full Text | Google Scholar

Hirano, T., Yasukawa, K., Harada, H., Taga, T., Watanabe, Y., Matsuda, T., et al. (1986). Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin. Nature 324, 73–76. doi: 10.1038/324073a0

PubMed Abstract | Crossref Full Text | Google Scholar

Hong, Y.-P., Lee, H.-C., and Kim, H.-T. (2015). Treadmill exercise after social isolation increases the levels of NGF, BDNF, and synapsin I to induce survival of neurons in the hippocampus, and improves depression-like behavior. J. Exerc. Nutr. Biochem. 19, 11–18. doi: 10.5717/jenb.2015.19.1.11

PubMed Abstract | Crossref Full Text | Google Scholar

Horiuchi, T., Mitoma, H., Harashima, S., Tsukamoto, H., and Shimoda, T. (2010). Transmembrane TNF-alpha: Structure, function and interaction with anti-TNF agents. Rheumatology 49, 1215–1228. doi: 10.1093/rheumatology/keq031

PubMed Abstract | Crossref Full Text | Google Scholar

Howells, D. W., Porritt, M. J., Wong, J. Y., Batchelor, P. E., Kalnins, R., Hughes, A. J., et al. (2000). Reduced BDNF mRNA expression in the Parkinson’s disease substantia nigra. Exp. Neurol. 166, 127–135. doi: 10.1006/exnr.2000.7483

PubMed Abstract | Crossref Full Text | Google Scholar

Huang, T., Larsen, K. T., Ried-Larsen, M., Møller, N. C., and Andersen, L. B. (2014). The effects of physical activity and exercise on brain-derived neurotrophic factor in healthy humans: A review. Scand. J. Med. Sci. Sports 24, 1–10. doi: 10.1111/sms.12069

PubMed Abstract | Crossref Full Text | Google Scholar

Huh, J. Y., Panagiotou, G., Mougios, V., Brinkoetter, M., Vamvini, M. T., Schneider, B. E., et al. (2012). FNDC5 and irisin in humans: I. predictors of circulating concentrations in serum and plasma and II. mRNA expression and circulating concentrations in response to weight loss and exercise. Metabolism 61, 1725–1738. doi: 10.1016/j.metabol.2012.09.002

PubMed Abstract | Crossref Full Text | Google Scholar

Ishii, T., Warabi, E., and Mann, G. E. (2019). Circadian control of BDNF-mediated Nrf2 activation in astrocytes protects dopaminergic neurons from ferroptosis. Free Radic. Biol. Med. 133, 169–178. doi: 10.1016/j.freeradbiomed.2018.09.002

PubMed Abstract | Crossref Full Text | Google Scholar

Islam, M. R., Valaris, S., Young, M. F., Haley, E. B., Luo, R., Bond, S. F., et al. (2021). Exercise hormone irisin is a critical regulator of cognitive function. Nat. Metab. 3, 1058–1070. doi: 10.1038/s42255-021-00438-z

PubMed Abstract | Crossref Full Text | Google Scholar

Izawa, S., Nishii, K., Aizu, N., Kito, T., Iwata, D., Chihara, T., et al. (2024). Effects of aerobic exercise and resistance training on cognitive function: Comparative study based on FNDC5/Irisin/BDNF pathway. Dement. Geriatr. Cogn. Disord. 53, 329–337. doi: 10.1159/000541093

PubMed Abstract | Crossref Full Text | Google Scholar

Jayanti, S., Moretti, R., Tiribelli, C., and Gazzin, S. (2022). Bilirubin prevents the TH+ dopaminergic neuron loss in a Parkinson’s disease model by acting on TNF-α. Int. J. Mol. Sci. 23:14276. doi: 10.3390/ijms232214276

PubMed Abstract | Crossref Full Text | Google Scholar

Jedrychowski, M. P., Wrann, C. D., Paulo, J. A., Gerber, K. K., Szpyt, J., Robinson, M. M., et al. (2015). Detection and quantitation of circulating human irisin by tandem mass spectrometry. Cell Metab. 22, 734–740. doi: 10.1016/j.cmet.2015.08.001

PubMed Abstract | Crossref Full Text | Google Scholar

Jeon, Y. K., and Ha, C. H. (2015). Expression of brain-derived neurotrophic factor, IGF-1 and cortisol elicited by regular aerobic exercise in adolescents. J. Phys. Ther. Sci. 27, 737–741. doi: 10.1589/jpts.27.737

PubMed Abstract | Crossref Full Text | Google Scholar

Jeong, D., Park, K., Lee, J., Choi, J., Du, H., Jeong, H., et al. (2024). Effects of resistance exercise and essential amino acid intake on muscle quality, myokine, and inflammation factors in young adult males. Nutrients 16:1688. doi: 10.3390/nu16111688

PubMed Abstract | Crossref Full Text | Google Scholar

Johansson, M. E., Cameron, I. G. M., Van der Kolk, N. M., de Vries, N. M., Klimars, E., Toni, I., et al. (2022). Aerobic exercise alters brain function and structure in Parkinson’s disease: A randomized controlled trial. Ann. Neurol. 91, 203–216. doi: 10.1002/ana.26291

PubMed Abstract | Crossref Full Text | Google Scholar

Kam, T. I., Park, H., Chou, S. C., Van Vranken, J. G., Mittenbühler, M. J., Kim, H., et al. (2022). Amelioration of pathologic α-synuclein-induced Parkinson’s disease by irisin. Proc. Natl. Acad. Sci. U. S. A. 119:e2204835119. doi: 10.1073/pnas.2204835119

PubMed Abstract | Crossref Full Text | Google Scholar

Kang, S. S., Zhang, Z., Liu, X., Manfredsson, F. P., Benskey, M. J., Cao, X., et al. (2017). TrkB neurotrophic activities are blocked by α-synuclein, triggering dopaminergic cell death in Parkinson’s disease. Proc. Natl. Acad. Sci. U. S. A. 114, 10773–10778. doi: 10.1073/pnas.1713969114

PubMed Abstract | Crossref Full Text | Google Scholar

Karakasis, C., Kalinderi, K., Katsarou, Z., Fidani, L., and Bostantjopoulou, S. (2011). Association of brain-derived neurotrophic factor (BDNF) Val66Met polymorphism with Parkinson’s disease in a Greek population. J. Clin. Neurosci. 18, 1744–1745. doi: 10.1016/j.jocn.2011.03.015

PubMed Abstract | Crossref Full Text | Google Scholar

Kaur, S., Bansal, Y., Kumar, R., and Bansal, G. (2020). A panoramic review of IL-6: Structure, pathophysiological roles and inhibitors. Bioorg. Med. Chem. 28:115327. doi: 10.1016/j.bmc.2020.115327

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, H., Wrann, C. D., Jedrychowski, M., Vidoni, S., Kitase, Y., Nagano, K., et al. (2018). Irisin mediates effects on bone and fat via αV integrin receptors. Cell 175, 1756–1768.e17. doi: 10.1016/j.cell.2018.10.025.

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, H. J., So, B., Choi, M., Kang, D., and Song, W. (2015). Resistance exercise training increases the expression of irisin concomitant with improvement of muscle function in aging mice and humans. Exp. Gerontol. 70, 11–17. doi: 10.1016/j.exger.2015.07.006

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, J. S., Taaffe, D. R., Galvão, D. A., Clay, T. D., Redfern, A. D., Hart, N. H., et al. (2023). Acute effect of high-intensity interval aerobic exercise on serum myokine levels and resulting tumour-suppressive effect in trained patients with advanced prostate cancer. Prostate Cancer Prostatic. Dis. 26, 795–801. doi: 10.1038/s41391-022-00624-4

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, T., Chang, J. S., Kim, H., Lee, K. H., and Kong, I. D. (2015). Intense walking exercise affects serum IGF-1 and IGFBP3. J. Lifestyle Med. 5, 21–25. doi: 10.15280/jlm.2015.5.1.21

PubMed Abstract | Crossref Full Text | Google Scholar

Kishimoto, T., and Ishizaka, K. (1973). Regulation of antibody response in vitro. VII. Enhancing soluble factors for IgG and IgE antibody response. J. Immunol. 111, 1194–1205.

Google Scholar

Klein, A. B., Williamson, R., Santini, M. A., Clemmensen, C., Ettrup, A., Rios, M., et al. (2011). Blood BDNF concentrations reflect brain-tissue BDNF levels across species. Int. J. Neuropsychopharmacol. 14, 347–353. doi: 10.1017/S1461145710000738

PubMed Abstract | Crossref Full Text | Google Scholar

Knaepen, K., Goekint, M., Heyman, E. M., and Meeusen, R. (2010). Neuroplasticity - exercise-induced response of peripheral brain-derived neurotrophic factor: A systematic review of experimental studies in human subjects. Sports Med. 40, 765–801. doi: 10.2165/11534530-000000000-00000

PubMed Abstract | Crossref Full Text | Google Scholar

Ko, B. J., Park, K. H., Shin, S., Zaichenko, L., Davis, C. R., Crowell, J. A., et al. (2016). Diet quality and diet patterns in relation to circulating cardiometabolic biomarkers. Clin. Nutr. 35, 484–490. doi: 10.1016/j.clnu.2015.03.022

PubMed Abstract | Crossref Full Text | Google Scholar

Koeda, T., Mavropalias, G., Mizumura, K., Katanosaka, K., and Nosaka, K. (2024). Changes in nerve growth factor in vastus lateralis muscle after the first versus second bout of one-leg eccentric cycling. Scand. J. Med. Sci. Sports 34:e14497. doi: 10.1111/sms.14497

PubMed Abstract | Crossref Full Text | Google Scholar

Koltun, K. J., Sterczala, A. J., Sekel, N. M., Krajewski, K. T., Martin, B. J., Lovalekar, M., et al. (2024). Effect of acute resistance exercise on bone turnover in young adults before and after concurrent resistance and interval training. Physiol. Rep. 12:e15906. doi: 10.14814/phy2.15906

PubMed Abstract | Crossref Full Text | Google Scholar

Kong, L., Miu, L., Yao, W., and Shi, Z. (2024). Effect of regular aerobic exercise on cognitive function, depression level and regulative role of neurotrophic factor: A prospective cohort study in the young and the middle-aged sample. Risk Manag. Healthc. Policy 17, 935–943. doi: 10.2147/RMHP.S456765

PubMed Abstract | Crossref Full Text | Google Scholar

Kozina, E., Byrne, M., and Smeyne, R. J. (2022). Mutant LRRK2 in lymphocytes regulates neurodegeneration via IL-6 in an inflammatory model of Parkinson’s disease. NPJ Parkinsons Dis. 8:24. doi: 10.1038/s41531-022-00289-9

PubMed Abstract | Crossref Full Text | Google Scholar

Kwok, J. Y. Y., Kwan, J. C. Y., Auyeung, M., Mok, V. C. T., Lau, C. K. Y., Choi, K. C., et al. (2019). Effects of mindfulness yoga vs stretching and resistance training exercises on anxiety and depression for people with parkinson disease: A randomized clinical trial. JAMA Neurol. 76, 755–763. doi: 10.1001/jamaneurol.2019.0534

PubMed Abstract | Crossref Full Text | Google Scholar

Lamichane, S., Baek, S. H., Kim, Y. J., Park, J. H., Dahal Lamichane, B., Jang, W. B., et al. (2019). MHY2233 attenuates replicative cellular senescence in human endothelial progenitor cells via SIRT1 signaling. Oxid. Med. Cell Longev. 2019:6492029. doi: 10.1155/2019/6492029

PubMed Abstract | Crossref Full Text | Google Scholar

Lan, G., Wang, P., Chan, R. B., Liu, Z., Yu, Z., Liu, X., et al. (2022). Astrocytic VEGFA: An essential mediator in blood-brain-barrier disruption in Parkinson’s disease. Glia 70, 337–353. doi: 10.1002/glia.24109

PubMed Abstract | Crossref Full Text | Google Scholar

Landry, T., Shookster, D., Chaves, A., Free, K., Nguyen, T., and Huang, H. (2022). Exercise increases NPY/AgRP and TH neuron activity in the hypothalamus of female mice. J. Endocrinol. 252, 167–177. doi: 10.1530/JOE-21-0250

PubMed Abstract | Crossref Full Text | Google Scholar

Lange, C., Storkebaum, E., de Almodóvar, C. R., Dewerchin, M., and Carmeliet, P. (2016). Vascular endothelial growth factor: A neurovascular target in neurological diseases. Nat. Rev. Neurol. 12, 439–454. doi: 10.1038/nrneurol.2016.88

PubMed Abstract | Crossref Full Text | Google Scholar

Lawrence, J. M., Schardien, K., Wigdahl, B., and Nonnemacher, M. R. (2023). Roles of neuropathology-associated reactive astrocytes: A systematic review. Acta Neuropathol. Commun. 11:42. doi: 10.1186/s40478-023-01526-9

PubMed Abstract | Crossref Full Text | Google Scholar

Lee, B., Shin, M., Park, Y., Won, S. Y., and Cho, K. S. (2021). Physical exercise-induced myokines in neurodegenerative diseases. Int. J. Mol. Sci. 22:5795. doi: 10.3390/ijms22115795

PubMed Abstract | Crossref Full Text | Google Scholar

Lee, P., Linderman, J. D., Smith, S., Brychta, R. J., Wang, J., Idelson, C., et al. (2014). Irisin and FGF21 are cold-induced endocrine activators of brown fat function in humans. Cell Metab. 19, 302–309. doi: 10.1016/j.cmet.2013.12.017

PubMed Abstract | Crossref Full Text | Google Scholar

Leem, Y. H., Park, J. S., Park, J. E., Kim, D. Y., and Kim, H. S. (2023). Suppression of neuroinflammation and α-synuclein oligomerization by rotarod walking exercise in subacute MPTP model of Parkinson’s disease. Neurochem. Int. 165:105519. doi: 10.1016/j.neuint.2023.105519

PubMed Abstract | Crossref Full Text | Google Scholar

Leite Silva, A. B. R., Gonçalves de Oliveira, R. W., Diógenes, G. P., de Castro Aguiar, M. F., Sallem, C. C., Lima, M. P. P., et al. (2023). Premotor, nonmotor and motor symptoms of Parkinson’s Disease: A new clinical state of the art. Ageing Res. Rev. 84:101834. doi: 10.1016/j.arr.2022.101834

PubMed Abstract | Crossref Full Text | Google Scholar

Levi-Montalcini, R. (1987). The nerve growth factor 35 years later. Science 237, 1154–1162. doi: 10.1126/science.3306916

PubMed Abstract | Crossref Full Text | Google Scholar

Li, B., Feng, L., Wu, X., Cai, M., Yu, J. J., and Tian, Z. (2022). Effects of different modes of exercise on skeletal muscle mass and function and IGF-1 signaling during early aging in mice. J. Exp. Biol. 225:jeb244650. doi: 10.1242/jeb.244650

PubMed Abstract | Crossref Full Text | Google Scholar

Li, H., Xiao, H., Lin, L., Jou, D., Kumari, V., Lin, J., et al. (2014). Drug design targeting protein-protein interactions (PPIs) using multiple ligand simultaneous docking (MLSD) and drug repositioning: Discovery of raloxifene and bazedoxifene as novel inhibitors of IL-6/GP130 interface. J. Med. Chem. 57, 632–641. doi: 10.1021/jm401144z

PubMed Abstract | Crossref Full Text | Google Scholar

Li, M., Li, J., Chen, H., and Zhu, M. (2023). VEGF-Expressing mesenchymal stem cell therapy for safe and effective treatment of pain in Parkinson’s disease. Cell Transplant 32:9636897221149130. doi: 10.1177/09636897221149130

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, L. F., Doherty, D. H., Lile, J. D., Bektesh, S., and Collins, F. (1993). GDNF: A glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 260, 1130–1132. doi: 10.1126/science.8493557

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, L. F., Zhang, T. J., Collins, F., and Armes, L. G. (1994). Purification and initial characterization of rat B49 glial cell line-derived neurotrophic factor. J. Neurochem. 63, 758–768. doi: 10.1046/j.1471-4159.1994.63020758.x

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, T. W., Tsai, S. F., and Kuo, Y. M. (2018). Physical exercise enhances neuroplasticity and delays Alzheimer’s disease. Brain Plast 4, 95–110. doi: 10.3233/BPL-180073

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, Y., Wang, J., Liu, X., Hu, Y., Zhang, Y., and Jiang, F. (2024). Synthesis, biological activity evaluation and mechanism analysis of new ganglioside GM3 derivatives as potential agents for nervous functional recovery. Eur. J. Med. Chem. 266:116108. doi: 10.1016/j.ejmech.2023.116108

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, J. L., and LeRoith, D. (1999). Insulin-like growth factor I is essential for postnatal growth in response to growth hormone. Endocrinology 140, 5178–5184. doi: 10.1210/endo.140.11.7151

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, L., Luo, L., Wei, J. A., Xu, X., So, K. F., and Zhang, L. (2024). Treadmill exercise reshapes cortical astrocytic and neuronal activity to improve motor learning deficits under chronic alcohol exposure. Neurosci. Bull. 40, 1287–1298. doi: 10.1007/s12264-024-01226-x

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, S., Du, F., Li, X., Wang, M., Duan, R., Zhang, J., et al. (2017). Effects and underlying mechanisms of irisin on the proliferation and apoptosis of pancreatic β cells. PLoS One 12:e0175498. doi: 10.1371/journal.pone.0175498

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, T. W., Chen, C. M., and Chang, K. H. (2022). Biomarker of neuroinflammation in Parkinson’s disease. Int. J. Mol. Sci. 23:4148. doi: 10.3390/ijms23084148

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, Y., Meng, X., Tang, C., Zheng, L., Tao, K., and Guo, W. (2024). Aerobic exercise modulates RIPK1-mediated MAP3K5/JNK and NF-κB pathways to suppress microglia activation and neuroinflammation in the hippocampus of D-gal-induced accelerated aging mice. Physiol. Behav. 286:114676. doi: 10.1016/j.physbeh.2024.114676

PubMed Abstract | Crossref Full Text | Google Scholar

Loprinzi, P. D., Day, S., and Deming, R. (2019). Acute exercise intensity and memory function: Evaluation of the transient hypofrontality hypothesis. Medicina 55:445. doi: 10.3390/medicina55080445

PubMed Abstract | Crossref Full Text | Google Scholar

Lotankar, S., Prabhavalkar, K. S., and Bhatt, L. K. (2017). Biomarkers for Parkinson’s disease: Recent advancement. Neurosci. Bull. 33, 585–597. doi: 10.1007/s12264-017-0183-5

PubMed Abstract | Crossref Full Text | Google Scholar

Ludtmann, M. H. R., Kostic, M., Horne, A., Gandhi, S., Sekler, I., and Abramov, A. Y. (2019). LRRK2 deficiency induced mitochondrial Ca2+ efflux inhibition can be rescued by Na+/Ca2+/Li+ exchanger upregulation. Cell Death Dis. 10:265. doi: 10.1038/s41419-019-1469-5

PubMed Abstract | Crossref Full Text | Google Scholar

Luo, D., Zhao, J., Cheng, Y., Lee, S. M., and Rong, J. (2018). N-Propargyl Caffeamide (PACA) ameliorates dopaminergic neuronal loss and motor dysfunctions in MPTP mouse model of Parkinson’s disease and in MPP+-Induced neurons via promoting the conversion of proNGF to NGF. Mol. Neurobiol. 55, 2258–2267. doi: 10.1007/s12035-017-0486-6

PubMed Abstract | Crossref Full Text | Google Scholar

Ma, J., Gao, J., Niu, M., Zhang, X., Wang, J., and Xie, A. (2020). P2X4R overexpression upregulates interleukin-6 and exacerbates 6-OHDA-Induced dopaminergic degeneration in a rat model of PD. Front. Aging Neurosci. 12:580068. doi: 10.3389/fnagi.2020.580068

PubMed Abstract | Crossref Full Text | Google Scholar

Mabeta, P., and Steenkamp, V. (2022). The VEGF/VEGFR axis revisited: Implications for cancer therapy. Int. J. Mol. Sci. 23:15585. doi: 10.3390/ijms232415585

PubMed Abstract | Crossref Full Text | Google Scholar

Macchi, Z. A., Koljack, C. E., Miyasaki, J. M., Katz, M., Galifianakis, N., Prizer, L. P., et al. (2020). Patient and caregiver characteristics associated with caregiver burden in Parkinson’s disease: A palliative care approach. Ann. Palliat. Med. 9, (Suppl. 1), S24–S33. doi: 10.21037/apm.2019.10.01

PubMed Abstract | Crossref Full Text | Google Scholar

Maes, M., Rachayon, M., Jirakran, K., Sodsai, P., and Sughondhabirom, A. (2023). Lower nerve growth factor levels in major depression and suicidal behaviors: Effects of adverse childhood experiences and recurrence of illness. Brain Sci. 13:1090. doi: 10.3390/brainsci13071090

PubMed Abstract | Crossref Full Text | Google Scholar

Mahalakshmi, B., Maurya, N., Lee, S. D., and Bharath Kumar, V. (2020). Possible neuroprotective mechanisms of physical exercise in neurodegeneration. Int. J. Mol. Sci. 21:5895. doi: 10.3390/ijms21165895

PubMed Abstract | Crossref Full Text | Google Scholar

Mahato, A. K., and Sidorova, Y. A. (2020). RET receptor tyrosine kinase: Role in neurodegeneration, obesity, and cancer. Int. J. Mol. Sci. 21:7108. doi: 10.3390/ijms21197108

PubMed Abstract | Crossref Full Text | Google Scholar

Majbour, N. K., Aasly, J. O., Hustad, E., Thomas, M. A., Vaikath, N. N., Elkum, N., et al. (2020). CSF total and oligomeric α-Synuclein along with TNF-α as risk biomarkers for Parkinson’s disease: A study in LRRK2 mutation carriers. Transl. Neurodegener. 9:15. doi: 10.1186/s40035-020-00192-4

PubMed Abstract | Crossref Full Text | Google Scholar

Malczynska-Sims, P., Chalimoniuk, M., Wronski, Z., Marusiak, J., and Sulek, A. (2022). High-intensity interval training modulates inflammatory response in Parkinson’s disease. Aging Clin. Exp. Res. 34, 2165–2176. doi: 10.1007/s40520-022-02153-5

PubMed Abstract | Crossref Full Text | Google Scholar

Martínez-Guardado, I., Arboleya, S., Grijota, F. J., Kaliszewska, A., Gueimonde, M., and Arias, N. (2022). The therapeutic role of exercise and probiotics in stressful brain conditions. Int. J. Mol. Sci. 23:3610. doi: 10.3390/ijms23073610

PubMed Abstract | Crossref Full Text | Google Scholar

Mata, I. F., Leverenz, J. B., Weintraub, D., Trojanowski, J. Q., Hurtig, H. I., Van Deerlin, V. M., et al. (2014). APOE, MAPT, and SNCA genes and cognitive performance in Parkinson disease. JAMA Neurol. 71, 1405–1412. doi: 10.1001/jamaneurol.2014.1455

PubMed Abstract | Crossref Full Text | Google Scholar

Mätlik, K., Garton, D. R., Montaño-Rodríguez, A. R., Olfat, S., Eren, F., Casserly, L., et al. (2022). Elevated endogenous GDNF induces altered dopamine signalling in mice and correlates with clinical severity in schizophrenia. Mol. Psychiatry 27, 3247–3261. doi: 10.1038/s41380-022-01554-2

PubMed Abstract | Crossref Full Text | Google Scholar

McAllister, A. K. (2001). Neurotrophins and neuronal differentiation in the central nervous system. Cell Mol. Life Sci. 58, 1054–1060. doi: 10.1007/PL00000920

PubMed Abstract | Crossref Full Text | Google Scholar

McCullough, M. J., Gyorkos, A. M., and Spitsbergen, J. M. (2013). Short-term exercise increases GDNF protein levels in the spinal cord of young and old rats. Neuroscience 240, 258–268. doi: 10.1016/j.neuroscience.2013.02.063

PubMed Abstract | Crossref Full Text | Google Scholar

McGlinchey, R. P., and Lee, J. C. (2015). Cysteine cathepsins are essential in lysosomal degradation of α-synuclein. Proc. Natl. Acad. Sci. U. S. A. 112, 9322–9327. doi: 10.1073/pnas.1500937112

PubMed Abstract | Crossref Full Text | Google Scholar

McMorris, T. (2016). Developing the catecholamines hypothesis for the acute exercise-cognition interaction in humans: Lessons from animal studies. Physiol. Behav. 165, 291–299. doi: 10.1016/j.physbeh.2016.08.011

PubMed Abstract | Crossref Full Text | Google Scholar

Mendes-Oliveira, J., Campos, F. L., Ferreira, S. A., Tomé, D., Fonseca, C. P., and Baltazar, G. (2023). Endogenous GDNF is unable to halt dopaminergic injury triggered by microglial activation. Cells 13:74. doi: 10.3390/cells13010074

PubMed Abstract | Crossref Full Text | Google Scholar

Mettu, P. S., Allingham, M. J., and Cousins, S. W. (2021). Incomplete response to Anti-VEGF therapy in neovascular AMD: Exploring disease mechanisms and therapeutic opportunities. Prog. Retin. Eye Res. 82:100906. doi: 10.1016/j.preteyeres.2020.100906

PubMed Abstract | Crossref Full Text | Google Scholar

Mihci, E., Ozkaynak, S. S., Sallakci, N., Kizilay, F., and Yavuzer, U. (2011). VEGF polymorphisms and serum VEGF levels in Parkinson’s disease. Neurosci. Lett. 494, 1–5. doi: 10.1016/j.neulet.2011.02.027

PubMed Abstract | Crossref Full Text | Google Scholar

Moghadasi, M., Akbari, F., and Najafi, P. (2024). Interaction of aerobic exercise and crocin improves memory, learning and hypocampic tau and neurotrophins gene expression in rats treated with trimethytin as a model of Alzheimer’s disease. Mol. Biol. Rep. 51:111. doi: 10.1007/s11033-023-09197-4

PubMed Abstract | Crossref Full Text | Google Scholar

Molska, M., Mruczyk, K., Cisek-Woźniak, A., Prokopowicz, W., Szydełko, P., Jakuszewska, Z., et al. (2024). The influence of intestinal microbiota on BDNF levels. Nutrients 16:2891. doi: 10.3390/nu16172891

PubMed Abstract | Crossref Full Text | Google Scholar

Moore, R. J., Owens, D. M., Stamp, G., Arnott, C., Burke, F., East, N., et al. (1999). Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis. Nat. Med. 5, 828–831. doi: 10.1038/10552

PubMed Abstract | Crossref Full Text | Google Scholar

Moriarty, N., Gantner, C. W., Hunt, C. P. J., Ermine, C. M., Frausin, S., Viventi, S., et al. (2022). A combined cell and gene therapy approach for homotopic reconstruction of midbrain dopamine pathways using human pluripotent stem cells. Cell Stem Cell 29, 434–448.e5. doi: 10.1016/j.stem.2022.01.013.

PubMed Abstract | Crossref Full Text | Google Scholar

Morland, C., Andersson, K. A., Haugen, ØP., Hadzic, A., Kleppa, L., Gille, A., et al. (2017). Exercise induces cerebral VEGF and angiogenesis via the lactate receptor HCAR1. Nat. Commun. 8:15557. doi: 10.1038/ncomms15557

PubMed Abstract | Crossref Full Text | Google Scholar

Mu, A., Wales, T. E., Zhou, H., Draga-Coletă, S. V., Gorgulla, C., Blackmore, K. A., et al. (2023). Irisin acts through its integrin receptor in a two-step process involving extracellular Hsp90α. Mol. Cell 83, 1903–1920.e12. doi: 10.1016/j.molcel.2023.05.008.

PubMed Abstract | Crossref Full Text | Google Scholar

Mu, L., Cai, J., Gu, B., Yu, L., Li, C., Liu, Q. S., et al. (2022). Treadmill exercise prevents decline in spatial learning and memory in 3×Tg-AD mice through enhancement of structural synaptic plasticity of the hippocampus and prefrontal cortex. Cells 11:244. doi: 10.3390/cells11020244

PubMed Abstract | Crossref Full Text | Google Scholar

Nicoletti, J. N., Shah, S. K., McCloskey, D. P., Goodman, J. H., Elkady, A., Atassi, H., et al. (2008). Vascular endothelial growth factor is up-regulated after status epilepticus and protects against seizure-induced neuronal loss in hippocampus. Neuroscience 151, 232–241. doi: 10.1016/j.neuroscience.2007.09.083

PubMed Abstract | Crossref Full Text | Google Scholar

Omer, N., Giladi, N., Gurevich, T., Bar-Shira, A., Gana-Weisz, M., Goldstein, O., et al. (2020). A possible modifying effect of the G2019S mutation in the LRRK2 gene on GBA Parkinson’s disease. Mov. Disord. 35, 1249–1253. doi: 10.1002/mds.28066

PubMed Abstract | Crossref Full Text | Google Scholar

Pahlavani, H. A. (2023). Exercise therapy to prevent and treat Alzheimer’s disease. Front. Aging Neurosci. 15:1243869. doi: 10.3389/fnagi.2023.1243869

PubMed Abstract | Crossref Full Text | Google Scholar

Palasz, E., Niewiadomski, W., Gasiorowska, A., Mietelska-Porowska, A., and Niewiadomska, G. (2019). Neuroplasticity and neuroprotective effect of treadmill training in the chronic mouse model of Parkinson’s disease. Neural Plast. 2019:8215017. doi: 10.1155/2019/8215017

PubMed Abstract | Crossref Full Text | Google Scholar

Palasz, E., Wysocka, A., Gasiorowska, A., Chalimoniuk, M., Niewiadomski, W., and Niewiadomska, G. (2020). BDNF as a promising therapeutic agent in Parkinson’s disease. Int. J. Mol. Sci. 21:1170. doi: 10.3390/ijms21031170

PubMed Abstract | Crossref Full Text | Google Scholar

Pan, W., Banks, W. A., Fasold, M. B., Bluth, J., and Kastin, A. J. (1998). Transport of brain-derived neurotrophic factor across the blood-brain barrier. Neuropharmacology 37, 1553–1561. doi: 10.1016/s0028-3908(98)00141-5

PubMed Abstract | Crossref Full Text | Google Scholar

Parkhurst, C. N., Yang, G., Ninan, I., Savas, J. N., Yates, J. R., Lafaille, J. J., et al. (2013). Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609. doi: 10.1016/j.cell.2013.11.030

PubMed Abstract | Crossref Full Text | Google Scholar

Patel, S., Homaei, A., El-Seedi, H. R., and Akhtar, N. (2018). Cathepsins: Proteases that are vital for survival but can also be fatal. Biomed. Pharmacother. 105, 526–532. doi: 10.1016/j.biopha.2018.05.148

PubMed Abstract | Crossref Full Text | Google Scholar

Patterson, S. L. (2015). Immune dysregulation and cognitive vulnerability in the aging brain: Interactions of microglia, IL-1β, BDNF and synaptic plasticity. Neuropharmacology 96, 11–18. doi: 10.1016/j.neuropharm.2014.12.020

PubMed Abstract | Crossref Full Text | Google Scholar

Pedersen, B. K. (2019). Physical activity and muscle-brain crosstalk. Nat. Rev. Endocrinol. 15, 383–392. doi: 10.1038/s41574-019-0174-x

PubMed Abstract | Crossref Full Text | Google Scholar

Pekkala, S., Wiklund, P. K., Hulmi, J. J., Ahtiainen, J. P., Horttanainen, M., Pöllänen, E., et al. (2013). Are skeletal muscle FNDC5 gene expression and irisin release regulated by exercise and related to health? J. Physiol. 591, 5393–5400. doi: 10.1113/jphysiol.2013.263707

PubMed Abstract | Crossref Full Text | Google Scholar

Perakakis, N., Triantafyllou, G. A., Fernández-Real, J. M., Huh, J. Y., Park, K. H., Seufert, J., et al. (2017). Physiology and role of irisin in glucose homeostasis. Nat. Rev. Endocrinol. 13, 324–337. doi: 10.1038/nrendo.2016.221

PubMed Abstract | Crossref Full Text | Google Scholar

Pignataro, P., Dicarlo, M., Zerlotin, R., Zecca, C., Dell’Abate, M. T., Buccoliero, C., et al. (2021). FNDC5/Irisin system in neuroinflammation and neurodegenerative diseases: Update and novel perspective. Int. J. Mol. Sci. 22:1605. doi: 10.3390/ijms22041605

PubMed Abstract | Crossref Full Text | Google Scholar

Pinkowska, A., Podhorska-Okołów, M., Dzięgiel, P., and Nowińska, K. (2021). The role of irisin in Cancer disease. Cells 10:1479. doi: 10.3390/cells10061479

PubMed Abstract | Crossref Full Text | Google Scholar

Piri, H., Sharifi, S., Nigjeh, S., and Haghdoost-Yazdi, H. (2022). Dopaminergic neuronal death in the substantia nigra associates with change in serum levels of TNF-α and IL-1β; Evidence from early experimental model of Parkinson’s disease. Neurol. Res. 44, 544–553. doi: 10.1080/01616412.2021.2024726

PubMed Abstract | Crossref Full Text | Google Scholar

Policastro, G., Brunelli, M., Tinazzi, M., Chiamulera, C., Emerich, D. F., and Paolone, G. (2020). Cytokine-, neurotrophin-, and motor rehabilitation-induced plasticity in Parkinson’s disease. Neural. Plast. 2020:8814028. doi: 10.1155/2020/8814028

PubMed Abstract | Crossref Full Text | Google Scholar

Polyzos, S. A., Kountouras, J., Anastasilakis, A. D., Geladari, E. V., and Mantzoros, C. S. (2014). Irisin in patients with nonalcoholic fatty liver disease. Metabolism 63, 207–217. doi: 10.1016/j.metabol.2013.09.013

PubMed Abstract | Crossref Full Text | Google Scholar

Pons-Espinal, M., Blasco-Agell, L., Fernandez-Carasa, I., Andrés-Benito, P., di Domenico, A., Richaud-Patin, Y., et al. (2024). Blocking IL-6 signaling prevents astrocyte-induced neurodegeneration in an iPSC-based model of Parkinson’s disease. JCI Insight. 9:e163359. doi: 10.1172/jci.insight.163359

PubMed Abstract | Crossref Full Text | Google Scholar

Qin, X. Y., Zhang, S. P., Cao, C., Loh, Y. P., and Cheng, Y. (2016). Aberrations in peripheral inflammatory cytokine levels in Parkinson disease: A systematic review and meta-analysis. JAMA Neurol. 73, 1316–1324. doi: 10.1001/jamaneurol.2016.2742

PubMed Abstract | Crossref Full Text | Google Scholar

Qiu, S., Cai, X., Sun, Z., Schumann, U., Zügel, M., and Steinacker, J. M. (2015). Chronic exercise training and circulating irisin in adults: A meta-analysis. Sports Med. 45, 1577–1588. doi: 10.1007/s40279-014-0293-4

PubMed Abstract | Crossref Full Text | Google Scholar

Qu, Y., Li, J., Qin, Q., Wang, D., Zhao, J., An, K., et al. (2023). A systematic review and meta-analysis of inflammatory biomarkers in Parkinson’s disease. NPJ Parkinsons Dis. 9:18. doi: 10.1038/s41531-023-00449-5

PubMed Abstract | Crossref Full Text | Google Scholar

Quesada, A., Lee, B. Y., and Micevych, P. E. (2008). PI3 kinase/Akt activation mediates estrogen and IGF-1 nigral DA neuronal neuroprotection against a unilateral rat model of Parkinson’s disease. Dev. Neurobiol. 68, 632–644. doi: 10.1002/dneu.20609

PubMed Abstract | Crossref Full Text | Google Scholar

Rabelo, P. C. R., Horta, N. A. C., Cordeiro, L. M. S., Poletini, M. O., Coimbra, C. C., Szawka, R. E., et al. (2017). Intrinsic exercise capacity in rats influences dopamine neuroplasticity induced by physical training. J. Appl. Physiol. 123, 1721–1729. doi: 10.1152/japplphysiol.00506.2017

PubMed Abstract | Crossref Full Text | Google Scholar

Rabiee, F., Lachinani, L., Ghaedi, S., Nasr-Esfahani, M. H., Megraw, T. L., and Ghaedi, K. (2020). New insights into the cellular activities of Fndc5/Irisin and its signaling pathways. Cell Biosci. 10:51. doi: 10.1186/s13578-020-00413-3

PubMed Abstract | Crossref Full Text | Google Scholar

Radak, Z., Zhao, Z., Koltai, E., Ohno, H., and Atalay, M. (2013). Oxygen consumption and usage during physical exercise: The balance between oxidative stress and ROS-dependent adaptive signaling. Antioxid. Redox Signal. 18, 1208–1246. doi: 10.1089/ars.2011.4498

PubMed Abstract | Crossref Full Text | Google Scholar

Rahmati, M., Keshvari, M., Xie, W., Yang, G., Jin, H., Li, H., et al. (2022). Resistance training and Urtica dioica increase neurotrophin levels and improve cognitive function by increasing age in the hippocampus of rats. Biomed. Pharmacother. 153:113306. doi: 10.1016/j.biopha.2022.113306

PubMed Abstract | Crossref Full Text | Google Scholar

Raschke, S., Elsen, M., Gassenhuber, H., Sommerfeld, M., Schwahn, U., Brockmann, B., et al. (2013). Evidence against a beneficial effect of irisin in humans. PLoS One 8:e73680. doi: 10.1371/journal.pone.0073680

PubMed Abstract | Crossref Full Text | Google Scholar

Requejo, C., Ruiz-Ortega, J. A., Bengoetxea, H., García-Blanco, A., Herrán, E., Aristieta, A., et al. (2017). Morphological changes in a severe model of Parkinson’s disease and its suitability to test the therapeutic effects of microencapsulated neurotrophic factors. Mol. Neurobiol. 54, 7722–7735. doi: 10.1007/s12035-016-0244-1

PubMed Abstract | Crossref Full Text | Google Scholar

Reycraft, J. T., Islam, H., Townsend, L. K., Hayward, G. C., Hazell, T. J., and Macpherson, R. E. K. (2020). Exercise intensity and recovery on circulating brain-derived neurotrophic factor. Med. Sci. Sports Exerc. 52, 1210–1217. doi: 10.1249/MSS.0000000000002242

PubMed Abstract | Crossref Full Text | Google Scholar

Ritz, B. R., and Paul, K. C. (2022). Recommending healthy diet and exercise to patients with parkinson disease-no reason to hold back. JAMA Netw. Open 5:e2227743. doi: 10.1001/jamanetworkopen.2022.27743

PubMed Abstract | Crossref Full Text | Google Scholar

Ross, O. A., Soto-Ortolaza, A. I., Heckman, M. G., Aasly, J. O., Abahuni, N., Annesi, G., et al. (2011). Association of LRRK2 exonic variants with susceptibility to Parkinson’s disease: A case-control study. Lancet Neurol. 10, 898–908. doi: 10.1016/S1474-4422(11)70175-2

PubMed Abstract | Crossref Full Text | Google Scholar

Ruan, Q., Zhang, L., Ruan, J., Zhang, X., Chen, J., Ma, C., et al. (2018). Detection and quantitation of irisin in human cerebrospinal fluid by tandem mass spectrometry. Peptides 103, 60–64. doi: 10.1016/j.peptides.2018.03.013

PubMed Abstract | Crossref Full Text | Google Scholar

Rui, Q., Ni, H., Li, D., Gao, R., and Chen, G. (2018). The role of LRRK2 in neurodegeneration of Parkinson disease. Curr. Neuropharmacol. 16, 1348–1357. doi: 10.2174/1570159X16666180222165418

PubMed Abstract | Crossref Full Text | Google Scholar

Ruiz-González, D., Hernández-Martínez, A., Valenzuela, P. L., Morales, J. S., and Soriano-Maldonado, A. (2021). Effects of physical exercise on plasma brain-derived neurotrophic factor in neurodegenerative disorders: A systematic review and meta-analysis of randomized controlled trials. Neurosci. Biobehav. Rev. 128, 394–405. doi: 10.1016/j.neubiorev.2021.05.025

PubMed Abstract | Crossref Full Text | Google Scholar

Russ, K., Teku, G., Bousset, L., Redeker, V., Piel, S., Savchenko, E., et al. (2021). TNF-α and α-synuclein fibrils differently regulate human astrocyte immune reactivity and impair mitochondrial respiration. Cell Rep. 34:108895. doi: 10.1016/j.celrep.2021.108895

PubMed Abstract | Crossref Full Text | Google Scholar

Sansone, L., Reali, V., Pellegrini, L., Villanova, L., Aventaggiato, M., Marfe, G., et al. (2013). SIRT1 silencing confers neuroprotection through IGF-1 pathway activation. J. Cell Physiol. 228, 1754–1761. doi: 10.1002/jcp.24334

PubMed Abstract | Crossref Full Text | Google Scholar

Sariola, H., and Saarma, M. (2003). Novel functions and signalling pathways for GDNF. J. Cell Sci. 116(Pt 19), 3855–3862. doi: 10.1242/jcs.00786

PubMed Abstract | Crossref Full Text | Google Scholar

Scheller, J., Chalaris, A., Schmidt-Arras, D., and Rose-John, S. (2011). The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 1813, 878–888. doi: 10.1016/j.bbamcr.2011.01.034

PubMed Abstract | Crossref Full Text | Google Scholar

Shahmoradian, S. H., Lewis, A. J., Genoud, C., Hench, J., Moors, T. E., Navarro, P. P., et al. (2019). Lewy pathology in Parkinson’s disease consists of crowded organelles and lipid membranes. Nat. Neurosci. 22, 1099–1109. doi: 10.1038/s41593-019-0423-2

PubMed Abstract | Crossref Full Text | Google Scholar

Shan, C., Zhang, C., and Zhang, C. (2024). The role of IL-6 in neurodegenerative disorders. Neurochem. Res. 49, 834–846. doi: 10.1007/s11064-023-04085-6

PubMed Abstract | Crossref Full Text | Google Scholar

Shang, X., Hao, X., Hou, W., Liu, J., Chi, R., Deng, X., et al. (2024). Exercise-induced modulation of myokine irisin on muscle-bone unit in the rat model of post-traumatic osteoarthritis. J. Orthop. Surg. Res. 19:49. doi: 10.1186/s13018-024-04532-2

PubMed Abstract | Crossref Full Text | Google Scholar

Shao, L., Li, H., Chen, J., Song, H., Zhang, Y., Wu, F., et al. (2017). Irisin suppresses the migration, proliferation, and invasion of lung cancer cells via inhibition of epithelial-to-mesenchymal transition. Biochem. Biophys. Res. Commun. 485, 598–605. doi: 10.1016/j.bbrc.2016.12.084

PubMed Abstract | Crossref Full Text | Google Scholar

Shastri, A., Bonifati, D. M., and Kishore, U. (2013). Innate immunity and neuroinflammation. Med. Inflamm. 2013:342931. doi: 10.1155/2013/342931

PubMed Abstract | Crossref Full Text | Google Scholar

Sheikh, M. A., Malik, Y. S., Xing, Z., Guo, Z., Tian, H., Zhu, X., et al. (2017). Polylysine-modified polyethylenimine (PEI-PLL) mediated VEGF gene delivery protects dopaminergic neurons in cell culture and in rat models of Parkinson’s Disease (PD). Acta Biomater. 54, 58–68. doi: 10.1016/j.actbio.2016.12.048

PubMed Abstract | Crossref Full Text | Google Scholar

Shi, X., Gu, Q., Fu, C., Ma, J., Li, D., Zheng, J., et al. (2024). Relationship of irisin with disease severity and dopamine uptake in Parkinson’s disease patients. Neuroimage Clin. 41:103555. doi: 10.1016/j.nicl.2023.103555

PubMed Abstract | Crossref Full Text | Google Scholar

Shi, X., Zheng, J., Ma, J., Li, D., Gu, Q., Chen, S., et al. (2023). Correlation between serum IGF-1 and EGF levels and neuropsychiatric and cognitive in Parkinson’s disease patients. Neurol. Sci. 44, 881–887. doi: 10.1007/s10072-022-06490-1

PubMed Abstract | Crossref Full Text | Google Scholar

Shin, K. O., Bae, J. Y., Woo, J., Jang, K. S., Kim, K. S., Park, J. S., et al. (2015). The effect of exercise on expression of myokine and angiogenesis mRNA in skeletal muscle of high fat diet induced obese rat. J. Exerc. Nutrit. Biochem. 19, 91–98. doi: 10.5717/jenb.2015.15061006

PubMed Abstract | Crossref Full Text | Google Scholar

Sidorova, Y. A., and Saarma, M. (2020). Small molecules and peptides targeting glial cell line-derived neurotrophic factor receptors for the treatment of neurodegeneration. Int. J. Mol. Sci. 21:6575. doi: 10.3390/ijms21186575

PubMed Abstract | Crossref Full Text | Google Scholar

Smith, A. D., and Zigmond, M. J. (2003). Can the brain be protected through exercise? Lessons from an animal model of parkinsonism. Exp. Neurol. 184, 31–39. doi: 10.1016/j.expneurol.2003.08.017

PubMed Abstract | Crossref Full Text | Google Scholar

Soori, R., Amini, A. A., Choobineh, S., Eskandari, A., Behjat, A., Ghram, A., et al. (2022). Exercise attenuates myocardial fibrosis and increases angiogenesis-related molecules in the myocardium of aged rats. Arch. Physiol. Biochem. 128, 1–6. doi: 10.1080/13813455.2019.1660370

PubMed Abstract | Crossref Full Text | Google Scholar

Sosero, Y. L., and Gan-Or, Z. (2023). LRRK2 and Parkinson’s disease: From genetics to targeted therapy. Ann. Clin. Transl. Neurol. 10, 850–864. doi: 10.1002/acn3.51776

PubMed Abstract | Crossref Full Text | Google Scholar

Stein, A. M., da Silva, T. M. V., Coelho, F. G. M., Rueda, A. V., Camarini, R., and Galduróz, R. F. S. (2021). Acute exercise increases circulating IGF-1 in Alzheimer’s disease patients, but not in older adults without dementia. Behav. Brain Res. 396:112903. doi: 10.1016/j.bbr.2020.112903

PubMed Abstract | Crossref Full Text | Google Scholar

Sterling, J. K., Kam, T. I., Guttha, S., Park, H., Baumann, B., Mehrabani-Tabari, A. A., et al. (2022). Interleukin-6 triggers toxic neuronal iron sequestration in response to pathological α-synuclein. Cell Rep. 38:110358. doi: 10.1016/j.celrep.2022.110358

PubMed Abstract | Crossref Full Text | Google Scholar

Stoka, V., Turk, V., and Turk, B. (2016). Lysosomal cathepsins and their regulation in aging and neurodegeneration. Ageing Res. Rev. 32, 22–37. doi: 10.1016/j.arr.2016.04.010

PubMed Abstract | Crossref Full Text | Google Scholar

Sujkowski, A., Hong, L., Wessells, R. J., and Todi, S. V. (2022). The protective role of exercise against age-related neurodegeneration. Ageing Res. Rev. 74:101543. doi: 10.1016/j.arr.2021.101543

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, M. F., Zhu, Y. L., Zhou, Z. L., Jia, X. B., Xu, Y. D., Yang, Q., et al. (2018). Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF-α signaling pathway. Brain Behav. Immun. 70, 48–60. doi: 10.1016/j.bbi.2018.02.005

PubMed Abstract | Crossref Full Text | Google Scholar

Szuhany, K. L., Bugatti, M., and Otto, M. W. (2015). A meta-analytic review of the effects of exercise on brain-derived neurotrophic factor. J. Psychiatr. Res. 60, 56–64. doi: 10.1016/j.jpsychires.2014.10.003

PubMed Abstract | Crossref Full Text | Google Scholar

Tabas, I., and Glass, C. K. (2013). Anti-inflammatory therapy in chronic disease: Challenges and opportunities. Science 339, 166–172. doi: 10.1126/science.1230720

PubMed Abstract | Crossref Full Text | Google Scholar

Tait, J. L., Duckham, R. L., Rantalainen, T., Milte, C. M., Main, L. C., Nowson, C. A., et al. (2024). Effects of a 6-month dual-task, power-based exercise program on cognitive function, neurological and inflammatory markers in older adults: Secondary analysis of a cluster randomised controlled trial. Geroscience 47, 1251–1268. doi: 10.1007/s11357-024-01316-8

PubMed Abstract | Crossref Full Text | Google Scholar

Takahashi, M., Ritz, J., and Cooper, G. M. (1985). Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell 42, 581–588. doi: 10.1016/0092-8674(85)90115-1

PubMed Abstract | Crossref Full Text | Google Scholar

Tang, C., Liu, M., Zhou, Z., Li, H., Yang, C., Yang, L., et al. (2023). Treadmill exercise alleviates cognition disorder by activating the FNDC5: Dual role of integrin αV/β5 in Parkinson’s disease. Int. J. Mol. Sci. 24:7830. doi: 10.3390/ijms24097830

PubMed Abstract | Crossref Full Text | Google Scholar

Tang, C. X., Chen, J., Shao, K. Q., Liu, Y. H., Zhou, X. Y., Ma, C. C., et al. (2023). Blunt dopamine transmission due to decreased GDNF in the PFC evokes cognitive impairment in Parkinson’s disease. Neural Regen. Res. 18, 1107–1117. doi: 10.4103/1673-5374.355816

PubMed Abstract | Crossref Full Text | Google Scholar

Tansey, M. G., Wallings, R. L., Houser, M. C., Herrick, M. K., Keating, C. E., and Joers, V. (2022). Inflammation and immune dysfunction in Parkinson disease. Nat. Rev. Immunol. 22, 657–673. doi: 10.1038/s41577-022-00684-6

PubMed Abstract | Crossref Full Text | Google Scholar

Testa, G., Cattaneo, A., and Capsoni, S. (2021). Understanding pain perception through genetic painlessness diseases: The role of NGF and proNGF. Pharmacol. Res. 169:105662. doi: 10.1016/j.phrs.2021.105662

PubMed Abstract | Crossref Full Text | Google Scholar

Thirupathi, A., Marqueze, L. F., Outeiro, T. F., Radak, Z., and Pinho, R. A. (2024). Physical exercise-induced activation of NRF2 and BDNF as a promising strategy for ferroptosis regulation in Parkinson’s disease. Neurochem. Res. 49, 1643–1654. doi: 10.1007/s11064-024-04152-6

PubMed Abstract | Crossref Full Text | Google Scholar

Thomas, G. (2002). Furin at the cutting edge: From protein traffic to embryogenesis and disease. Nat. Rev. Mol. Cell Biol. 3, 753–766. doi: 10.1038/nrm934

PubMed Abstract | Crossref Full Text | Google Scholar

Tian, J., Yu, T., Xu, Y., Pu, S., Lv, Y., Zhang, X., et al. (2018). Swimming training reduces neuroma pain by regulating neurotrophins. Med. Sci. Sports Exerc. 50, 54–61. doi: 10.1249/MSS.0000000000001411

PubMed Abstract | Crossref Full Text | Google Scholar

Tolosa, E., Garrido, A., Scholz, S. W., and Poewe, W. (2021). Challenges in the diagnosis of Parkinson’s disease. Lancet Neurol. 20, 385–397. doi: 10.1016/S1474-4422(21)00030-2

PubMed Abstract | Crossref Full Text | Google Scholar

Tommasini, E., Missaglia, S., Vago, P., Galvani, C., Pecci, C., Rampinini, E., et al. (2024). The time course of irisin release after an acute exercise: Relevant implications for health and future experimental designs. Eur. J. Transl. Myol. 34:12693. doi: 10.4081/ejtm.2024.12693

PubMed Abstract | Crossref Full Text | Google Scholar

Tran, A. P., and Silver, J. (2021). Cathepsins in neuronal plasticity. Neural Regen. Res. 16, 26–35. doi: 10.4103/1673-5374.286948

PubMed Abstract | Crossref Full Text | Google Scholar

Tsai, C. L., Pan, C. Y., Tseng, Y. T., Chen, F. C., Chang, Y. C., and Wang, T. C. (2021). Acute effects of high-intensity interval training and moderate-intensity continuous exercise on BDNF and irisin levels and neurocognitive performance in late middle-aged and older adults. Behav. Brain Res. 413:113472. doi: 10.1016/j.bbr.2021.113472

PubMed Abstract | Crossref Full Text | Google Scholar

Tsukita, K., Sakamaki-Tsukita, H., and Takahashi, R. (2022). Long-term effect of regular physical activity and exercise habits in patients with early Parkinson disease. Neurology 98, e859–e871. doi: 10.1212/WNL.0000000000013218

PubMed Abstract | Crossref Full Text | Google Scholar

Tung, Y. T., Liao, Y. C., Yeh, T. H., Tsao, S. P., Chang, C. C., Shih, W. T., et al. (2024). 10 weeks low intensity treadmill exercise intervention ameliorates motor deficits and sustains muscle mass via decreasing oxidative damage and increasing mitochondria function in a rat model of Parkinson’s disease. Life Sci. 350:122733. doi: 10.1016/j.lfs.2024.122733

PubMed Abstract | Crossref Full Text | Google Scholar

Tuon, T., Souza, P. S., Santos, M. F., Pereira, F. T., Pedroso, G. S., Luciano, T. F., et al. (2015). Physical training regulates mitochondrial parameters and neuroinflammatory mechanisms in an experimental model of Parkinson’s disease. Oxid. Med. Cell Longev. 2015:261809. doi: 10.1155/2015/261809

PubMed Abstract | Crossref Full Text | Google Scholar

Turk, V., Stoka, V., Vasiljeva, O., Renko, M., Sun, T., Turk, B., et al. (2012). Cysteine cathepsins: From structure, function and regulation to new frontiers. Biochim. Biophys. Acta 1824, 68–88. doi: 10.1016/j.bbapap.2011.10.002

PubMed Abstract | Crossref Full Text | Google Scholar

van Praag, H., Shubert, T., Zhao, C., and Gage, F. H. (2005). Exercise enhances learning and hippocampal neurogenesis in aged mice. J. Neurosci. 25, 8680–8685. doi: 10.1523/JNEUROSCI.1731-05.2005

PubMed Abstract | Crossref Full Text | Google Scholar

Venditti, S., Verdone, L., Pesce, C., Tocci, N., Caserta, M., and Ben-Soussan, T. D. (2015). Creating Well-Being: Increased creativity and proNGF decrease following quadrato motor training. Biomed. Res. Int. 2015:275062. doi: 10.1155/2015/275062

PubMed Abstract | Crossref Full Text | Google Scholar

Vilela, T. C., Muller, A. P., Damiani, A. P., Macan, T. P., da Silva, S., Canteiro, P. B., et al. (2017). Strength and aerobic exercises improve spatial memory in aging rats through stimulating distinct neuroplasticity mechanisms. Mol. Neurobiol. 54, 7928–7937. doi: 10.1007/s12035-016-0272-x

PubMed Abstract | Crossref Full Text | Google Scholar

Vints, W. A. J., Šeikinaitė, J., Gökçe, E., Kušleikienė, S., Šarkinaite, M., Valatkeviciene, K., et al. (2024). Resistance exercise effects on hippocampus subfield volumes and biomarkers of neuroplasticity and neuroinflammation in older adults with low and high risk of mild cognitive impairment: A randomized controlled trial. Geroscience 46, 3971–3991. doi: 10.1007/s11357-024-01110-6

PubMed Abstract | Crossref Full Text | Google Scholar

Vitt, U. A., Hsu, S. Y., and Hsueh, A. J. (2001). Evolution and classification of cystine knot-containing hormones and related extracellular signaling molecules. Mol. Endocrinol. 15, 681–694. doi: 10.1210/mend.15.5.0639

PubMed Abstract | Crossref Full Text | Google Scholar

Wagner, G., Herbsleb, M., de la Cruz, F., Schumann, A., Brünner, F., Schachtzabel, C., et al. (2015). Hippocampal structure, metabolism, and inflammatory response after a 6-week intense aerobic exercise in healthy young adults: A controlled trial. J. Cereb. Blood Flow Metab. 35, 1570–1578. doi: 10.1038/jcbfm.2015.125

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, J., Zhu, Q., Wang, Y., Peng, J., Shao, L., and Li, X. (2022). Irisin protects against sepsis-associated encephalopathy by suppressing ferroptosis via activation of the Nrf2/GPX4 signal axis. Free Radic. Biol. Med. 187, 171–184. doi: 10.1016/j.freeradbiomed.2022.05.023

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, R., and Holsinger, R. M. D. (2018). Exercise-induced brain-derived neurotrophic factor expression: Therapeutic implications for Alzheimer’s dementia. Ageing Res. Rev. 48, 109–121. doi: 10.1016/j.arr.2018.10.002

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, T. H., Feng, Z. T., Wei, P., Li, H., Shi, Z. J., and Li, L. Y. (2008). Effects of pcDNA3-beta-NGF gene-modified BMSC on the rat model of Parkinson’s disease. J. Mol. Neurosci. 35, 161–169. doi: 10.1007/s12031-007-9032-8

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Y., Liang, J., Xu, B., Yang, J., Wu, Z., and Cheng, L. (2024). TrkB/BDNF signaling pathway and its small molecular agonists in CNS injury. Life Sci. 336:122282. doi: 10.1016/j.lfs.2023.122282

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Y., Pu, Z., Zhang, Y., Du, Z., Guo, Z., and Bai, Q. (2023). Exercise training has a protective effect in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice model with improved neural and intestinal pathology and modified intestinal flora. Behav. Brain Res. 439:114240. doi: 10.1016/j.bbr.2022.114240

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Y. H., Luo, D. L., Jiang, H. C., Nie, Z. B., Shao, L., and Qi, H. X. (2022). Effects of aerobic exercise on inflammatory factors in healthy adults: A meta-analysis. Eur. Rev. Med. Pharmacol. Sci. 26, 4163–4175. doi: 10.26355/eurrev_202206_29053

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Z. (2021). Regulation of cell cycle progression by growth factor-induced cell signaling. Cells 10:3327. doi: 10.3390/cells10123327

PubMed Abstract | Crossref Full Text | Google Scholar

Weaver, S. R., Skinner, B. D., Furlong, R., Lucas, R. A. I., Cable, N. T., Rendeiro, C., et al. (2021). Cerebral hemodynamic and neurotrophic factor responses are dependent on the type of exercise. Front. Physiol. 11:609935. doi: 10.3389/fphys.2020.609935

PubMed Abstract | Crossref Full Text | Google Scholar

Xie, L., Huang, R., Liu, S., Wu, W., Su, A., Li, R., et al. (2019). A positive feedback loop of SIRT1 and miR17HG promotes the repair of DNA double-stranded breaks. Cell Cycle 18, 2110–2123. doi: 10.1080/15384101.2019.1641388

PubMed Abstract | Crossref Full Text | Google Scholar

Yadati, T., Houben, T., Bitorina, A., and Shiri-Sverdlov, R. (2020). The ins and outs of cathepsins: Physiological function and role in disease management. Cells 9:1679. doi: 10.3390/cells9071679

PubMed Abstract | Crossref Full Text | Google Scholar

Yakovlev, A. A., and Gulyaeva, N. V. (2015). Possible role of proteases in preconditioning of brain cells to pathological conditions. Biochemistry 80, 163–171. doi: 10.1134/S0006297915020030.

PubMed Abstract | Crossref Full Text | Google Scholar

Yarrow, J. F., White, L. J., McCoy, S. C., and Borst, S. E. (2010). Training augments resistance exercise induced elevation of circulating brain derived neurotrophic factor (BDNF). Neurosci. Lett. 479, 161–165. doi: 10.1016/j.neulet.2010.05.058

PubMed Abstract | Crossref Full Text | Google Scholar

Yu, M., King, B., Ewert, E., Su, X., Mardiyati, N., Zhao, Z., et al. (2016). Exercise activates p53 and negatively regulates IGF-1 pathway in epidermis within a skin cancer model. PLoS One 11:e0160939. doi: 10.1371/journal.pone.0160939

PubMed Abstract | Crossref Full Text | Google Scholar

Zang, Q., Wang, S., Qi, Y., Zhang, L., Huang, C., Xiu, Y., et al. (2023). Running exercise improves spatial learning and memory ability and enhances angiogenesis in the cerebral cortex via endogenous nitric oxide. Behav. Brain Res. 439:114243. doi: 10.1016/j.bbr.2022.114243

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Q., Xiang, S., Chen, X., Rong, Y., Huang, L., Chen, Z., et al. (2024). Irisin attenuates acute glaucoma-induced neuroinflammation by activating microglia-integrin αVβ5/AMPK and promoting autophagy. Int. Immunopharmacol. 138:112545. doi: 10.1016/j.intimp.2024.112545

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, S., Fan, Y., Cao, X., Deng, C., Xu, J., Zhou, Q., et al. (2024). Treadmill exercise improves cerebral ischemia injury by regulating microglia polarization via downregulation of MMP12. Int. Immunopharmacol. 142(Pt B):113210. doi: 10.1016/j.intimp.2024.113210

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, T., Liu, W., Bai, Q., and Gao, S. (2023). The therapeutic effects of yoga in people with Parkinson’s disease: A mini-review. Ann. Med. 55:2294935. doi: 10.1080/07853890.2023.2294935

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, X., Molsberry, S. A., Schwarzschild, M. A., Ascherio, A., and Gao, X. (2022). Association of diet and physical activity with all-cause mortality among adults with Parkinson disease. JAMA Netw. Open 5:e2227738. doi: 10.1001/jamanetworkopen.2022.27738

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, X., Xu, S., Hu, Y., Liu, Q., Liu, C., Chai, H., et al. (2023). Irisin exhibits neuroprotection by preventing mitochondrial damage in Parkinson’s disease. NPJ Parkinsons Dis. 9:13. doi: 10.1038/s41531-023-00453-9

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Gao, X., Bai, X., Yao, S., Chang, Y. Z., and Gao, G. (2022). The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl. Neurodegener. 11:39. doi: 10.1186/s40035-022-00313-1

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Li, R., Meng, Y., Li, S., Donelan, W., Zhao, Y., et al. (2014). Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes 63, 514–525. doi: 10.2337/db13-1106

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, C., Wu, Y., Zhu, S., Liu, H., and Xu, S. (2024). Irisin protects musculoskeletal homeostasis via a mitochondrial quality control mechanism. Int. J. Mol. Sci. 25:10116. doi: 10.3390/ijms251810116

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, Y., Haney, M. J., Jin, Y. S., Uvarov, O., Vinod, N., Lee, Y. Z., et al. (2019). GDNF-expressing macrophages restore motor functions at a severe late-stage, and produce long-term neuroprotective effects at an early-stage of Parkinson’s disease in transgenic Parkin Q311X(A) mice. J. Control. Release 315, 139–149. doi: 10.1016/j.jconrel.2019.10.027

PubMed Abstract | Crossref Full Text | Google Scholar

Zheng, S. Y., Li, H. X., Xu, R. C., Miao, W. T., Dai, M. Y., Ding, S. T., et al. (2021). Potential roles of gut microbiota and microbial metabolites in Parkinson’s disease. Ageing Res. Rev. 69:101347. doi: 10.1016/j.arr.2021.101347

PubMed Abstract | Crossref Full Text | Google Scholar

Zhong, Y., Liu, M. M., Li, J. C., Lu, T. C., Cao, X., Yang, Y. J., et al. (2024). In vitro drug screening models derived from different PC12 cell lines for exploring Parkinson’s disease based on electrochemical signals of catecholamine neurotransmitters. Mikrochim Acta 191:170. doi: 10.1007/s00604-024-06250-2

PubMed Abstract | Crossref Full Text | Google Scholar

Zoladz, J. A., Majerczak, J., Zeligowska, E., Mencel, J., Jaskolski, A., Jaskolska, A., et al. (2014). Moderate-intensity interval training increases serum brain-derived neurotrophic factor level and decreases inflammation in Parkinson’s disease patients. J. Physiol. Pharmacol. 65, 441–448.

Google Scholar

Keywords: Parkinson’s disease, exercise, cytokines, irisin, BDNF, TNF-α

Citation: Li W-Q, Yang J-H, Liu L-L, Ding S-T, Yu B, Jiang L, Yan N and Liu H-D (2025) Exercise as a multitarget therapy: modulating myokines, neurotrophins, and inflammation in Parkinson’s disease. Front. Aging Neurosci. 17:1580029. doi: 10.3389/fnagi.2025.1580029

Received: 20 February 2025; Accepted: 15 August 2025;
Published: 26 August 2025.

Edited by:

Shigeki Arawaka, Osaka Medical and Pharmaceutical University Faculty of Medicine, Japan

Reviewed by:

Margaret E. Rice, New York University, United States
Sok Kean Khoo, Grand Valley State University, United States

Copyright © 2025 Li, Yang, Liu, Ding, Yu, Jiang, Yan and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Han-Deng Liu, aGRsaXVAY3FtdS5lZHUuY24=

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.