ArticlePublisher preview available

The biphasic activity of autophagy and heat shock protein response in peripheral blood mononuclear cells following acute resistance exercise in resistance-trained males

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Purpose Autophagy and heat shock protein (HSP) response are proteostatic systems involved in the acute and adaptive responses to exercise. These systems may upregulate sequentially following cellular stress including acute exercise, however, currently few data exist in humans. This study investigated the autophagic and HSP responses to acute intense lower body resistance exercise in peripheral blood mononuclear cells (PBMCs) with and without branched-chain amino acids (BCAA) supplementation. Methods Twenty resistance-trained males (22.3 ± 1.5 yr; 175.4 ± .7 cm; 86.4 ± 15.6 kg) performed a bout of intense lower body resistance exercise and markers of autophagy and HSP70 were measured immediately post- (IPE) and 2, 4, 24, 48, and 72 h post-exercise. Prior to resistance exercise, 10 subjects were randomly assigned to BCAA supplementation of 0.22 g/kg/d for 5 days pre-exercise and up to 72 h following exercise while the other 10 subjects consumed a placebo (PLCB). Results There were no difference in autophagy markers or HSP70 expression between BCAA and PLCB groups. LC3II protein expression was significantly lower 2 and 4 h post-exercise compared to pre-exercise. LC3II: I ratio was not different at any time point compared to pre-exercise. Protein expression of p62 was lower IPE, 2, and 4 h post-exercise and elevated 24 h post-exercise. HSP70 expression was elevated 48 and 72 h post-exercise. Conclusions Autophagy and HSP70 are upregulated in PBMCs following intense resistance exercise with autophagy increasing initially post-exercise and HSP response in the latter period. Moreover, BCAA supplementation did not affect this response. Graphical Abstract
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
European Journal of Applied Physiology (2024) 124:2981–2992
https://doi.org/10.1007/s00421-024-05503-5
ORIGINAL ARTICLE
The biphasic activity ofautophagy andheat shock protein response
inperipheral blood mononuclear cells followingacute resistance
exercise inresistance‑trained males
KurtA.Escobar1 · TrishaA.VanDusseldorp2,3· KellyE.Johnson4· MatthewStratton5· JamesJ.McCormick6·
TerenceMoriarity7· KarolDokladny9· RogerA.Vaughan10· ChadM.Kerksick11· LenKravitz8· ChristineM.Mermier8
Received: 9 January 2024 / Accepted: 25 April 2024 / Published online: 21 May 2024
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2024, corrected publication 2024
Abstract
Purpose Autophagy and heat shock protein (HSP) response are proteostatic systems involved in the acute and adaptive
responses to exercise. These systems may upregulate sequentially following cellular stress including acute exercise, however,
currently few data exist in humans. This study investigated the autophagic and HSP responses to acute intense lower body
resistance exercise in peripheral blood mononuclear cells (PBMCs) with and without branched-chain amino acids (BCAA)
supplementation.
Methods Twenty resistance-trained males (22.3 ± 1.5yr; 175.4 ± .7cm; 86.4 ± 15.6kg) performed a bout of intense lower
body resistance exercise and markers of autophagy and HSP70 were measured immediately post- (IPE) and 2, 4, 24, 48, and
72h post-exercise. Prior to resistance exercise, 10 subjects were randomly assigned to BCAA supplementation of 0.22g/
kg/d for 5days pre-exercise and up to 72h following exercise while the other 10 subjects consumed a placebo (PLCB).
Results There were no difference in autophagy markers or HSP70 expression between BCAA and PLCB groups. LC3II
protein expression was significantly lower 2 and 4h post-exercise compared to pre-exercise. LC3II: I ratio was not different
at any time point compared to pre-exercise. Protein expression of p62 was lower IPE, 2, and 4h post-exercise and elevated
24h post-exercise. HSP70 expression was elevated 48 and 72h post-exercise.
Conclusions Autophagy and HSP70 are upregulated in PBMCs following intense resistance exercise with autophagy increas-
ing initially post-exercise and HSP response in the latter period. Moreover, BCAA supplementation did not affect this
response.
Communicated by William J. Kraemer.
* Kurt A. Escobar
kurt.escobar@csulb.edu
1 Physiology ofSport andExercise Lab, Department
ofKinesiology, California State University, Long Beach,
LongBeach, CA, USA
2 Bonafide Health, LLC p/b JDS Therapeutics, Harrison, NY,
USA
3 Department ofHealth andExercise Sciences, Jacksonville
University, Jacksonville, FL, USA
4 Department ofKinesiology, Coastal Carolina University,
Conway, SC, USA
5 Department ofHealth, Kinesiology, andSport, University
ofSouth Alabama, Mobile, AL, USA
6 Human andEnvironmental Physiology Research Unit, School
ofHuman Kinetics, University ofOttawa, Ottawa, Canada
7 Department ofKinesiology, University ofNorthern Iowa,
CedarFalls, USA
8 Department ofHealth, Exercise, andSport Science,
University ofNew Mexico, Albuquerque, NM, USA
9 Department ofInternal Medicine, Health Sciences Center,
University ofNew Mexico, Albuquerque, NM, USA
10 Department ofExercise Science, Congdon School ofHealth
Sciences, High Point University, HighPoint, NC, USA
11 Exercise andPerformance Nutrition Laboratory, College
ofScience, Technology, andHealth, Lindenwood University,
St.Charles, MO, USA
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Article
Full-text available
The heat shock factor 1 (HSF1)-mediated stress response pathway and autophagy processes play important roles in the maintenance of proteostasis. Autophagy processes are subdivided into three subtypes: macroautophagy, chaperone-mediated autophagy (CMA), and microautophagy. Recently, molecular chaperones and co-factors were shown to be involved in the selective degradation of substrates by these three autophagy processes. This evidence suggests that autophagy processes are regulated in a coordinated manner by the HSF1-mediated stress response pathway. Recently, various studies have demonstrated that proteostasis pathways including HSF1 and autophagy are implicated in longevity. Furthermore, they serve as therapeutic targets for aging-related diseases such as cancer and neurodegenerative diseases. In the future, these studies will underpin the development of therapies against various diseases.
Article
Full-text available
Background Macroautophagy/autophagy is an essential recycling process that is involved in a wide range of biological functions as well as in diseases. The regulation of autophagy by exercise and the associated health benefits have been revealed by rodent studies over the past decade, but the evidence from human studies remains inconclusive. Methods The MEDLINE, Embase, Cochrane, Scopus, and Web of Science databases were systematically searched from inception until September 2022. Human studies that explored potential effects of physical exercise on autophagy at the protein level were selected according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. A random-effects model was used for the meta-analysis. Results Twenty-six studies were included in the meta-analysis. Subgroup analyses revealed that an acute bout of resistance exercise attenuated autophagy, as characterized by lower levels of microtubule-associated proteins 1A/1B light chain 3B (LC3-II) and higher levels of sequestosome 1 (SQSTM1). In contrast, the long-term resistance exercise elevated autophagy, as shown by higher levels of LC3-II and lower levels of SQSTM1. No significant changes in LC3-II levels were observed with moderate- or vigorous-intensity endurance exercise either as an acute bout or long-term. In terms of tissue types, exercise exerted opposite effects between skeletal muscles and peripheral blood mononuclear cells (PBMCs), whereby autophagy was suppressed in skeletal muscles when activated in the PBMCs. Other meta-analyses have also shown significant alterations in the level of many canonical autophagic and mitophagic proteins, including unc-51 like autophagy activating kinase (ULK1)S317, ULK1S757, Beclin-1, ATG12, BCL2/adenovirus E1B 19 kDa protein-interacting protein 3, and PARKIN following exercise, suggesting the activation of canonical autophagy and mitophagy, although the scope of those analyses was more limited. Conclusion Our findings demonstrate that physical exercise probably regulates autophagy in an exercise modality- and tissue-dependent manner in humans, although further investigation is needed. Customized exercise prescriptions should be aimed for when implementing exercise to regulate autophagy in humans. Abbreviations: ATG: autophagy-related gene; BCL2L13: BCL2-like 13; BECN1: beclin1; BNIP3: BCL2/adenovirus E1B 19 kDa protein-interacting protein 3; GABARAP: gamma-aminobutyric acid receptor-associated protein; GAPDH: glyceraldehyde 3-phosphate dehydrogenase; LAMP2: lysosome-associated membrane protein 2; LC3B: microtubule-associated proteins 1A/1B light chain 3B; MD: mean difference; mTOR: mammalian target of rapamycin; PBMC: peripheral blood mononuclear cells; PINK1: PTEN-induced kinase 1; PRISMA: preferred reporting items for systematic review and meta-analysis; SD: standard deviation; SQSTM1: sequestosome 1; ULK1: unc-51 like autophagy activating kinase 1; VDAC1: voltage-dependent anion-selective channel 1.
Article
Full-text available
Branched-chain amino acids (BCAAs) are oxidized in the muscle and result in stimulating anabolic signals—which in return may optimize performance, body composition and recovery. Meanwhile, among athletes, the evidence about BCAA supplementation is not clear. The aim of this study was to review the effects of BCAAs in athletic populations. The research was conducted in three databases: Web of Science (all databases), PubMed and Scopus. The inclusion criteria involved participants classified both as athletes and people who train regularly, and who were orally supplemented with BCAAs. The risk of bias was individually assessed for each study using the revised Cochrane risk of bias tool for randomized trials (RoB 2.0). From the 2298 records found, 24 studies met the inclusion criteria. Although BCAAs tended to activate anabolic signals, the benefits on performance and body composition were negligible. On the other hand, studies that included resistance participants showed that BCAAs attenuated muscle soreness after exercise, while in endurance sports the findings were inconsistent. The protocols of BCAA supplements differed considerably between studies. Moreover, most of the studies did not report the total protein intake across the day and, consequently, the benefits of BCAAs should be interpreted with caution.
Article
Full-text available
Immunometabolism has been the focus of extensive research over the last years, especially in terms of augmenting anti-tumor immune responses. Regulatory T cells (Tregs) are a subset of CD4⁺ T cells, which have been known for their immunosuppressive roles in various conditions including anti-tumor immune responses. Even though several studies aimed to target Tregs in the tumor microenvironment (TME), such approaches generally result in the inhibition of the Tregs non-specifically, which may cause immunopathologies such as autoimmunity. Therefore, specifically targeting the Tregs in the TME would be vital in terms of achieving a successful and specific treatment. Recently, an association between Tregs and isoleucine, which represents one type of branched-chain amino acids (BCAAs), has been demonstrated. The presence of isoleucine seems to affect majorly Tregs, rather than conventional T cells. Considering the fact that Tregs bear several distinct metabolic features in the TME, targeting their immunometabolic pathways may be a rational approach. In this Review, we provide a general overview on the potential distinct metabolic features of T cells, especially focusing on BCAAs in Tregs as well as in their subtypes.
Article
Autophagy is a vital cellular process, essential to maintaining cellular function during acute physiological stressors including exercise and heat stress. We previously showed that autophagy occurs during exercise in an intensity-dependent manner in peripheral blood mononuclear cells (PBMCs) from young men, with elevated responses in the heat. However, given autophagy declines with age, it is unclear whether a similar pattern of response occurs in older adults. Therefore, we evaluated autophagy and the cellular stress response (i.e., apoptosis, inflammation, and the heat shock response [HSR]) in PBMCs from 10 healthy older men (mean [SD]: aged 70 years [5]) in response to 30 minutes of semi-recumbent cycling at low-, moderate-, and vigorous-intensities (40, 55, and 70% maximal oxygen consumption (VO 2max ), respectively) in a temperate (25°C) environment, with an additional vigorous-intensity bout (70% of VO 2max ) performed in a hot environment (40°C). Responses were evaluated before and after exercise, as well as throughout a 6-hour seated recovery period performed in the same environmental conditions as the respective exercise bout. Proteins were assessed via Western blot. While we observed elevations in mean body temperature with each increase in exercise intensity, autophagy was only stimulated during vigorous-intensity exercise, where we observed elevations in LC3-II (p<0.05). However, when the same exercise was performed in the heat, the LC3-II response was attenuated, which was accompanied by significant p62 accumulation (p<0.05). Altogether, our findings demonstrate that older adults exhibit autophagic impairments when the same vigorous-intensity exercise is performed in hot environments, potentially underlying heat-induced cellular vulnerability in older men.
Article
Muscle mass is balanced between hypertrophy and atrophy by cellular processes, including activation of the Akt-mTOR signaling cascade. Stressors apart from exercise and nutrition, such as heat stress, can stimulate the heat shock protein A (HSPA) and C (HSPC) families alongside hypertrophic signaling factors and muscle growth. The effects of heat stress on HSP expression and Akt-mTOR activation in human skeletal muscle, and their magnitude of activation compared to known hypertrophic stimuli is unclear. Here we show a single session of whole-body heat stress followed by resistance exercise increases the expression of HSPA as well as activation of the Akt-mTOR cascade in skeletal muscle compared to resistance exercise in a healthy, resistance trained population. Heat stress alone may also exert similar effects, though the responses are notably variable and require further investigation. Additionally, acute heat stress in C2C12 muscle cells enhanced myotube growth and myogenic fusion, albeit to a lesser degree than growth factor mediated hypertrophy. Though the mechanisms by which heat stress stimulates hypertrophy related signaling and the potential mechanistic role of HSPs remain unclear, these findings provide additional evidence implicating heat stress as a novel growth stimulus when combined with resistance exercise in human skeletal muscle and alone in isolated murine muscle cells. We believe these findings will help drive further applied and mechanistic investigation into how heat stress influences muscular hypertrophy and atrophy.
Article
Peripheral blood mononuclear cells (PBMCs) are a variety of specialized immune cells produced in the bone marrow from hematopoietic stem cells (HSCs) that work together to protect our bodies from harmful pathogens. From a metabolic point of view, these cells can serve as sentinel tissue source for distinguishing multiple types of whole-body physiological perturbations. The significant interaction of PBMCs with systemic physiology makes these cells an attractive target for several interventions such as physical exercise. Analyses of oxidative/inflammatory and metabolic markers of PBMCs obtained from unhealthy and healthy humans have been used in monitoring immune response in different exercise conditions. It is already a common consensus that regular practice of physical exercise, that is planned, structured, and repetitive, influences personal health by altering the metabolic state and the immune system. However, the role of distinct metabolic processes responsible for maintaining metabolic balance during physical exercise in PBMCs is not fully understood. Furthermore, a complete dose-response analysis between different exercise protocols and biomarkers capable of predicting physical performance needs to be better elucidated. The absence of published reviews on this topic compromises the understanding of the crosstalk between the metabolic adaptations of PBMCs and exercise-induced changes in the immune system. Given the above, this review highlights the main findings in the literature involving the responses of PBMCs in the inflammatory/oxidative stress induced by physical exercise. The present review also highlights how distinct phenotypes and functional diversity of PBMCs make these cells an accessible alternative for assessing exercise-induced metabolic adaptations.
Article
Branched-chain amino acids (BCAAs) provide nutrient signals for cell survival and growth. How BCAAs affect CD8+ T cell functions remains unexplored. Herein, we report that accumulation of BCAAs in CD8+ T cells due to the impairment of BCAA degradation in 2C-type serine/threonine protein phosphatase (PP2Cm)-deficient mice leads to hyper-activity of CD8+ T cells and enhanced anti-tumor immunity. CD8+ T cells from PP2Cm-/- mice upregulate glucose transporter Glut1 expression in a FoxO1-dependent manner with more glucose uptake, as well as increased glycolysis and oxidative phosphorylation. Moreover, BCAA supplementation recapitulates CD8+ T cell hyper-functions and synergizes with anti-PD-1, in line with a better prognosis in NSCLC patients containing high BCAAs when receiving anti-PD-1 therapy. Our finding thus reveals that accumulation of BCAAs promotes effector function and anti-tumor immunity of CD8+ T cells through reprogramming glucose metabolism, making BCAAs alternative supplementary components to increase the clinical efficacy of anti-PD-1 immunotherapy against tumors.
Article
Autophagy is essential to maintaining cellular homeostasis in all eukaryotic cells and to tolerance of acute stressors such as starvation, heat, and recovery following exercise. Limited information exists regarding the exercise intensity-dependent autophagic response in humans, and it is unknown how environmental heat stress may modulate this response. Therefore, we evaluated autophagy and accompanying pathways of cellular stress (the heat shock response [HSR], apoptosis, and acute inflammation) in peripheral blood mononuclear cells (PBMCs) from 10 young men (mean [SD]; 22 [2] years) before, immediately after and up to 6h post-exercise recovery from 30 minutes of low-, moderate-, and high-intensity semi-recumbent cycling (40, 55 and 70% of maximal oxygen consumption (VO 2max ), respectively) in a temperate environment (25°C) and at 70% of VO 2max in a hot environment (40°C). Changes in protein content were analyzed via Western blot. Each increase in exercise intensity was associated with elevations in mean body temperature. LC3-II increased following moderate-intensity exercise, with further increases following high-intensity exercise (p < 0.05). However, an increase in beclin-2 and ULK1, with a decrease in p62 was only observed after high-intensity exercise, which was paralleled by elevated TNF-α and cleaved-caspase-3, with the HSR peaking at 6h after exercise (p < 0.05). When exercise was performed in the heat, greater LC3-II and cleaved-caspase-3 accumulation was observed, however beclin-2 declined in recovery (p < 0.05). Therefore, our findings indicate that autophagy in PBMCs during exercise may be associated with greater heat strain exhibited during increasing exercise intensities, which is modulated by exposure to heat.
Article
Background: Resistance exercise is beneficial for the immune system, including decreased susceptibility to infections and improved effectiveness of vaccinations. This review aims to provide a systematic analysis of the literature regarding the impact of resistance exercise on immune cells in the blood circulation. Materials and methods: The protocol of this review followed the PRISMA guidelines and registered in PROSPERO (ID: CRD42020157834). PubMed and Web-of-Science were systematically searched for relevant articles. Outcomes were divided into two categories: 1) inflammatory gene expression or secretion of inflammation-related cytokines and 2) other aspects such as cell migration, proliferation, apoptosis, phagocytosis, and redox status. Results: Thirty intervention studies were included in this review, of which 11 articles were randomized controlled trials and six non-randomized controlled trials. Although only resistance exercise interventions were included, there was a high heterogeneity regarding specific exercise modalities. The most frequently studied outcome measures were the gene and protein expression levels in peripheral blood mononuclear cells (PBMC). This review reveals that already one acute exercise bout activates the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway in PBMC. Although resistance exercise induces an acute cytosolic oxidative stress response, the antioxidant enzyme expression is improved after resistance training period. Natural killer cell activity increases in older but decreases in younger adults immediately after a resistance exercise bout. Moreover, resistance exercise improves neutrophil phagocytic activity. Finally, effects on lymphocyte proliferation remain unclear. Conclusions: The results of this systematic review demonstrate that resistance exercise has beneficial effects on several aspects of immune cell function both in young and older individuals. Acute changes in immune cell function occur already after a single bout of resistance exercise. However, regular resistance training during several weeks seems necessary to obtain beneficial adaptations that can be related to better immunity and reduced inflammation. The effects documented in this review confirm the beneficial effects of resistance exercise in young as well as older persons on the immune cell function.