1
|
Pradana F, Nijjar T, Cox PA, Morgan PT, Podlogar T, Lucas SJE, Drayson MT, Kinsella FAM, Wadley AJ. Brief cycling intervals incrementally increase the number of hematopoietic stem and progenitor cells in human peripheral blood. Front Physiol 2024; 15:1327269. [PMID: 39139483 PMCID: PMC11319260 DOI: 10.3389/fphys.2024.1327269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction Peripheral blood stem cell (PBSC) donation is the primary procedure used to collect hematopoietic stem and progenitor cells (HSPCs) for hematopoietic stem cell transplantation. Single bouts of exercise transiently enrich peripheral blood with HSPCs and cytolytic natural killer cells (CD56dim), which are important in preventing post-transplant complications. To provide a rationale to investigate the utility of exercise in a PBSC donation setting (≈3 h), this study aimed to establish whether interval cycling increased peripheral blood HSPC and CD56dim concentrations to a greater degree than continuous cycling. Methods In a randomised crossover study design, eleven males (mean ± SD: age 25 ± 7 years) undertook bouts of moderate intensity continuous exercise [MICE, 30 min, 65%-70% maximum heart rate (HRmax)], high-volume high intensity interval exercise (HV-HIIE, 4 × 4 min, 80%-85% HRmax) and low-volume HIIE (LV-HIIE, 4 × 2 min, 90%-95% HRmax). The cumulative impact of each interval on circulating HSPC (CD34+CD45dimSSClow) and CD56dim concentrations (cells/µL), and the bone marrow homing potential of HSPCs (expression of CXCR-4 and VLA-4) were determined. Results There was an increase in HSPC concentration after two intervals of LV-HIIE (Rest: 1.84 ± 1.55 vs. Interval 2: 2.94 ± 1.34, P = 0.01) and three intervals of HV-HIIE only (Rest: 2.05 ± 0.86 vs. Interval 3: 2.51 ± 1.05, P = 0.04). The concentration of all leukocyte subsets increased after each trial, with this greatest for CD56dim NK cells, and in HIIE vs. MICE (LV-HIIE: 4.77 ± 2.82, HV-HIIE: 4.65 ± 2.06, MICE: 2.44 ± 0.77, P < 0.0001). These patterns were observed for concentration, not frequency of CXCR-4+ and VLA-4+ HSPCs, which was unaltered. There was a marginal decrease in VLA-4, but not CXCR-4 expression on exercise-mobilised HSPCs after all trials (P < 0.0001). Discussion The results of the present study indicate that HIIE caused a more marked increase in HSPC and CD56dim NK cell concentrations than MICE, with mobilised HSPCs maintaining their bone marrow homing phenotype. LV-HIIE evoked an increase in HSPC concentration after just 2 × 2-minute intervals. The feasibility and clinical utility of interval cycling in a PBSC donation context should therefore be evaluated.
Collapse
Affiliation(s)
- Fendi Pradana
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
- Nutrition Study Program, Faculty of Public Health, Tadulako University, Palu, Indonesia
| | - Tarondeep Nijjar
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Phoebe A. Cox
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul T. Morgan
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Tim Podlogar
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Samuel J. E. Lucas
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Mark T. Drayson
- Clinical Immunology Service, University of Birmingham, Birmingham, United Kingdom
| | - Francesca A. M. Kinsella
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Alex J. Wadley
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
2
|
Gollie JM, Ryan AS, Sen S, Patel SS, Kokkinos PF, Harris-Love MO, Scholten JD, Blackman MR. Exercise for patients with chronic kidney disease: from cells to systems to function. Am J Physiol Renal Physiol 2024; 326:F420-F437. [PMID: 38205546 PMCID: PMC11208028 DOI: 10.1152/ajprenal.00302.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic kidney disease (CKD) is among the leading causes of death and disability, affecting an estimated 800 million adults globally. The underlying pathophysiology of CKD is complex creating challenges to its management. Primary risk factors for the development and progression of CKD include diabetes mellitus, hypertension, age, obesity, diet, inflammation, and physical inactivity. The high prevalence of diabetes and hypertension in patients with CKD increases the risk for secondary consequences such as cardiovascular disease and peripheral neuropathy. Moreover, the increased prevalence of obesity and chronic levels of systemic inflammation in CKD have downstream effects on critical cellular functions regulating homeostasis. The combination of these factors results in the deterioration of health and functional capacity in those living with CKD. Exercise offers protective benefits for the maintenance of health and function with age, even in the presence of CKD. Despite accumulating data supporting the implementation of exercise for the promotion of health and function in patients with CKD, a thorough description of the responses and adaptations to exercise at the cellular, system, and whole body levels is currently lacking. Therefore, the purpose of this review is to provide an up-to-date comprehensive review of the effects of exercise training on vascular endothelial progenitor cells at the cellular level; cardiovascular, musculoskeletal, and neural factors at the system level; and physical function, frailty, and fatigability at the whole body level in patients with CKD.
Collapse
Affiliation(s)
- Jared M Gollie
- Research and Development Service, Washington DC Veterans Affairs Medical Center, Washington, District of Columbia, United States
- Department of Health, Human Function, and Rehabilitation Sciences, The George Washington University, Washington, District of Columbia, United States
| | - Alice S Ryan
- Department of Medicine, University of Maryland, Baltimore, Maryland, United States
- Division of Geriatrics and Palliative Medicine, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, United States
| | - Sabyasachi Sen
- Department of Medicine, Washington DC Veterans Affairs, Medical Center, Washington, District of Columbia, United States
- Department of Medicine, The George Washington University, Washington, District of Columbia, United States
| | - Samir S Patel
- Research and Development Service, Washington DC Veterans Affairs Medical Center, Washington, District of Columbia, United States
- Department of Medicine, Washington DC Veterans Affairs, Medical Center, Washington, District of Columbia, United States
- Department of Medicine, The George Washington University, Washington, District of Columbia, United States
| | - Peter F Kokkinos
- Division of Cardiology, Washington DC Veterans Affairs Medical Center, Washington, District of Columbia, United States
- Department of Kinesiology and Health, Rutgers University, New Brunswick, New Jersey, United States
| | - Michael O Harris-Love
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Geriatric Research Education and Clinical Center, Eastern Colorado Veterans Affairs Health Care System, Denver, Colorado, United States
| | - Joel D Scholten
- Physical Medicine and Rehabilitation Service, Washington DC Veterans Affairs Medical Center, Washington, District of Columbia, United States
| | - Marc R Blackman
- Research and Development Service, Washington DC Veterans Affairs Medical Center, Washington, District of Columbia, United States
- Department of Medicine, Washington DC Veterans Affairs, Medical Center, Washington, District of Columbia, United States
- Department of Medicine, The George Washington University, Washington, District of Columbia, United States
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
- Department of Rehabilitation Medicine, Georgetown University, Washington, District of Columbia, United States
| |
Collapse
|
3
|
Saheli M, Moshrefi M, Baghalishahi M, Mohkami A, Firouzi Y, Suzuki K, Khoramipour K. Cognitive Fitness: Harnessing the Strength of Exerkines for Aging and Metabolic Challenges. Sports (Basel) 2024; 12:57. [PMID: 38393277 PMCID: PMC10891799 DOI: 10.3390/sports12020057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Addressing cognitive impairment (CI) represents a significant global challenge in health and social care. Evidence suggests that aging and metabolic disorders increase the risk of CI, yet promisingly, physical exercise has been identified as a potential ameliorative factor. Specifically, there is a growing understanding that exercise-induced cognitive improvement may be mediated by molecules known as exerkines. This review delves into the potential impact of aging and metabolic disorders on CI, elucidating the mechanisms through which various exerkines may bolster cognitive function in this context. Additionally, the discussion extends to the role of exerkines in facilitating stem cell mobilization, offering a potential avenue for improving cognitive impairment.
Collapse
Affiliation(s)
- Mona Saheli
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran; (M.S.); (M.B.)
| | - Mandana Moshrefi
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran;
| | - Masoumeh Baghalishahi
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran; (M.S.); (M.B.)
| | - Amirhossein Mohkami
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar 9617976487, Iran;
| | - Yaser Firouzi
- Department of Exercise Physiology, Faculty of Sport Sciences, Shahid Bahonar University, Kerman 7616913439, Iran;
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| | - Kayvan Khoramipour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran
| |
Collapse
|
4
|
Papadopetraki A, Giannopoulos A, Maridaki M, Zagouri F, Droufakou S, Koutsilieris M, Philippou A. The Role of Exercise in Cancer-Related Sarcopenia and Sarcopenic Obesity. Cancers (Basel) 2023; 15:5856. [PMID: 38136400 PMCID: PMC10741686 DOI: 10.3390/cancers15245856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
One of the most common adverse effects of cancer and its therapeutic strategies is sarcopenia, a condition which is characterised by excess muscle wasting and muscle strength loss due to the disrupted muscle homeostasis. Moreover, cancer-related sarcopenia may be combined with the increased deposition of fat mass, a syndrome called cancer-associated sarcopenic obesity. Both clinical conditions have significant clinical importance and can predict disease progression and survival. A growing body of evidence supports the claim that physical exercise is a safe and effective complementary therapy for oncology patients which can limit the cancer- and its treatment-related muscle catabolism and promote the maintenance of muscle mass. Moreover, even after the onset of sarcopenia, exercise interventions can counterbalance the muscle mass loss and improve the clinical appearance and quality of life of cancer patients. The aim of this narrative review was to describe the various pathophysiological mechanisms, such as protein synthesis, mitochondrial function, inflammatory response, and the hypothalamic-pituitary-adrenal axis, which are regulated by exercise and contribute to the management of sarcopenia and sarcopenic obesity. Moreover, myokines, factors produced by and released from exercising muscles, are being discussed as they appear to play an important role in mediating the beneficial effects of exercise against sarcopenia.
Collapse
Affiliation(s)
- Argyro Papadopetraki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.)
| | - Antonios Giannopoulos
- Section of Sports Medicine, Department of Community Medicine & Rehabilitation, Umeå University, 901 87 Umeå, Sweden;
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University, Leicestershire LE11 3TU, UK
| | - Maria Maridaki
- Faculty of Physical Education and Sport Science, National and Kapodistrian University of Athens, 172 37 Dafne, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | | | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.)
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (A.P.)
| |
Collapse
|
5
|
Rao A, Gupta A, Kain V, Halade GV. Extrinsic and intrinsic modulators of inflammation-resolution signaling in heart failure. Am J Physiol Heart Circ Physiol 2023; 325:H433-H448. [PMID: 37417877 PMCID: PMC10538986 DOI: 10.1152/ajpheart.00276.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Chronic and uncleared inflammation is the root cause of various cardiovascular diseases. Fundamentally, acute inflammation is supportive when overlapping with safe clearance of inflammation termed resolution; however, if the lifestyle-directed extrinsic factors such as diet, sleep, exercise, or physical activity are misaligned, that results in unresolved inflammation. Although genetics play a critical role in cardiovascular health, four extrinsic risk factors-unhealthy processed diet, sleep disruption or fragmentation, sedentary lifestyle, thereby, subsequent stress-have been identified as heterogeneous and polygenic triggers of heart failure (HF), which can result in several complications with indications of chronic inflammation. Extrinsic risk factors directly impact endogenous intrinsic factors, such as using fatty acids by immune-responsive enzymes [lipoxygenases (LOXs)/cyclooxygenases (COXs)/cytochromes-P450 (CYP450)] to form resolution mediators that activate specific resolution receptors. Thus, the balance of extrinsic factors such as diet, sleep, and physical activity feed-forward the coordination of intrinsic factors such as fatty acids-enzymes-bioactive lipid receptors that modulates the immune defense, metabolic health, inflammation-resolution signaling, and cardiac health. Future research on lifestyle- and aging-associated molecular patterns is warranted in the context of intrinsic and extrinsic factors, immune fitness, inflammation-resolution signaling, and cardiac health.
Collapse
Affiliation(s)
- Archana Rao
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| | - Akul Gupta
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Internal Medicine, Heart Institute, University of South Florida, Tampa, Florida, United States
| |
Collapse
|
6
|
Ross M, Kargl CK, Ferguson R, Gavin TP, Hellsten Y. Exercise-induced skeletal muscle angiogenesis: impact of age, sex, angiocrines and cellular mediators. Eur J Appl Physiol 2023:10.1007/s00421-022-05128-6. [PMID: 36715739 DOI: 10.1007/s00421-022-05128-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/25/2022] [Indexed: 01/31/2023]
Abstract
Exercise-induced skeletal muscle angiogenesis is a well-known physiological adaptation that occurs in humans in response to exercise training and can lead to endurance performance benefits, as well as improvements in cardiovascular and skeletal tissue health. An increase in capillary density in skeletal muscle improves diffusive oxygen exchange and waste extraction, and thus greater fatigue resistance, which has application to athletes but also to the general population. Exercise-induced angiogenesis can significantly contribute to improvements in cardiovascular and metabolic health, such as the increase in muscle glucose uptake, important for the prevention of diabetes. Recently, our understanding of the mechanisms by which angiogenesis occurs with exercise has grown substantially. This review will detail the biochemical, cellular and biomechanical signals for exercise-induced skeletal muscle angiogenesis, including recent work on extracellular vesicles and circulating angiogenic cells. In addition, the influence of age, sex, exercise intensity/duration, as well as recent observations with the use of blood flow restricted exercise, will also be discussed in detail. This review will provide academics and practitioners with mechanistic and applied evidence for optimising training interventions to promote physical performance through manipulating capillarisation in skeletal muscle.
Collapse
Affiliation(s)
- Mark Ross
- School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, Scotland, UK.
| | - Christopher K Kargl
- Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, USA.,Department of Health and Kinesiology, Max E. Wastl Human Performance Laboratory, Purdue University, West Lafayette, USA
| | - Richard Ferguson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Timothy P Gavin
- Department of Health and Kinesiology, Max E. Wastl Human Performance Laboratory, Purdue University, West Lafayette, USA
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Molecular mechanisms of exercise contributing to tissue regeneration. Signal Transduct Target Ther 2022; 7:383. [PMID: 36446784 PMCID: PMC9709153 DOI: 10.1038/s41392-022-01233-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Physical activity has been known as an essential element to promote human health for centuries. Thus, exercise intervention is encouraged to battle against sedentary lifestyle. Recent rapid advances in molecular biotechnology have demonstrated that both endurance and resistance exercise training, two traditional types of exercise, trigger a series of physiological responses, unraveling the mechanisms of exercise regulating on the human body. Therefore, exercise has been expected as a candidate approach of alleviating a wide range of diseases, such as metabolic diseases, neurodegenerative disorders, tumors, and cardiovascular diseases. In particular, the capacity of exercise to promote tissue regeneration has attracted the attention of many researchers in recent decades. Since most adult human organs have a weak regenerative capacity, it is currently a key challenge in regenerative medicine to improve the efficiency of tissue regeneration. As research progresses, exercise-induced tissue regeneration seems to provide a novel approach for fighting against injury or senescence, establishing strong theoretical basis for more and more "exercise mimetics." These drugs are acting as the pharmaceutical alternatives of those individuals who cannot experience the benefits of exercise. Here, we comprehensively provide a description of the benefits of exercise on tissue regeneration in diverse organs, mainly focusing on musculoskeletal system, cardiovascular system, and nervous system. We also discuss the underlying molecular mechanisms associated with the regenerative effects of exercise and emerging therapeutic exercise mimetics for regeneration, as well as the associated opportunities and challenges. We aim to describe an integrated perspective on the current advances of distinct physiological mechanisms associated with exercise-induced tissue regeneration on various organs and facilitate the development of drugs that mimics the benefits of exercise.
Collapse
|
8
|
Vanhie JJ, Kim W, Ek Orloff L, Ngu M, Collao N, De Lisio M. The role of exercise-and high fat diet-induced bone marrow extracellular vesicles in stress hematopoiesis. Front Physiol 2022; 13:1054463. [PMID: 36505084 PMCID: PMC9728614 DOI: 10.3389/fphys.2022.1054463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Exercise and obesity regulate hematopoiesis, in part through alterations in cellular and soluble components of the bone marrow niche. Extracellular vesicles (EVs) are components of the bone marrow niche that regulate hematopoiesis; however, the role of exercise training or obesity induced EVs in regulating hematopoiesis remains unknown. To address this gap, donor EVs were isolated from control diet-fed, sedentary mice (CON-SED), control diet-fed exercise trained mice (CON-EX), high fat diet-fed, sedentary mice (HFD-SED), and high fat diet-fed, exercise trained mice (HFD-EX) and injected into recipient mice undergoing stress hematopoiesis. Hematopoietic and niche cell populations were quantified, and EV miRNA cargo was evaluated. EV content did not differ between the four groups. Mice receiving HFD-EX EVs had fewer hematopoietic stem cells (HSCs) (p < 0.01), long-term HSC (p < 0.05), multipotent progenitors (p < 0.01), common myeloid progenitors (p<0.01), common lymphoid progenitors (p < 0.01), and granulocyte-macrophage progenitors (p < 0.05), compared to mice receiving HFD-SED EVs. Similarly, mice receiving EX EVs had fewer osteoprogenitor cells compared to SED (p < 0.05) but enhanced mesenchymal stromal cell (MSC) osteogenic differentiation in vitro (p < 0.05) compared to SED EVs. HFD EVs enhanced mesenchymal stromal cell (MSC) adipogenesis in vitro (p < 0.01) compared to CON EVs. HFD-EX EVs had lower microRNA-193 and microRNA-331-5p content, microRNAs implicated in inhibiting osteogenesis and leukemic cell expansion respectively, compared to HFD-SED EVs. The results identify alterations in EV cargo as a novel mechanism by which exercise training alters stress hematopoiesis and the bone marrow niche.
Collapse
Affiliation(s)
- James J. Vanhie
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Wooseok Kim
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Lisa Ek Orloff
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Matthew Ngu
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Nicolas Collao
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Michael De Lisio
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada,*Correspondence: Michael De Lisio,
| |
Collapse
|
9
|
Huang J, Pu Y, Xu K, Ding Q, Sun R, Yin L, Zhang J, Pu Y. High expression of HIF-1α alleviates benzene-induced hematopoietic toxicity and immunosuppression in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 311:119928. [PMID: 35970343 DOI: 10.1016/j.envpol.2022.119928] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/15/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Benzene exposure can cause pancytopenia and immunosuppression, leading to serious diseases such as aplastic anemia (AA) or acute myeloid leukemia (AML), but the underlying mechanism has not been fully elucidated. Hypoxia-inducible factor 1 (HIF-1) is an important transcription factor that regulates many downstream target genes. In this study, we reported a novel mechanism by which high expression of HIF-1α alleviated benzene toxicity. Mice with high expression of HIF-1α (HIF-1α+) were obtained by the Tet-on system and doxycycline induction, and they and wild-type (WT) mice were exposed to 150 mg/kg benzene for 0, 1, 3, 7, 10, 14, and 28 days. Dynamic changes in hematopoietic and immune-related indicators and the role of HIF-1α were explored. The level of white blood cells in mice reached the highest level on the third day, and immunity was activated and then suppressed within 10 days. Significant pancytopenia and immunosuppression occurred at 14 days and were more pronounced at 28 days. The levels of HIF-1α, EPO, VEGF, RORγt, and IL-17 in WT mice gradually decreased with increasing benzene exposure days, while the levels of Foxp3 and IL-10 increased. These changes were alleviated in HIF-1α+ mice. High expression of HIF-1α increased the levels of EPO and VEGF, which helped to maintain the stability of the hematopoietic microenvironment. Simultaneously, it attenuated benzene-induced immunosuppression by alleviating the Th17/Treg imbalance. HIF-1α is expected to be a new target for benzene-induced diseases such as AA and AML.
Collapse
Affiliation(s)
- Jiawei Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yunqiu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Kai Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Qin Ding
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Rongli Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
10
|
Abstract
Lifestyle factors are modifiable behavioral factors that have a significant impact on health and longevity. Diet-induced obesity and physical activity/exercise are two prevalent lifestyle factors that have strong relationships to overall health. The mechanisms linking obesity to negative health outcomes and the mechanisms linking increased participation in physical activity/exercise to positive health outcomes are beginning to be elucidated. Chronic inflammation, due in part to overproduction of myeloid cells from hematopoietic stem cells (HSCs) in the bone marrow, is an established mechanism responsible for the negative health effects of obesity. Recent work has shown that exercise training can reverse the aberrant myelopoiesis present in obesity in part by restoring the bone marrow microenvironment. Specifically, exercise training reduces marrow adipose tissue, increases HSC retention factor expression, and reduces pro-inflammatory cytokine levels in the bone marrow. Other, novel mechanistic factors responsible for these exercise-induced effects, including intercellular communication using extracellular vesicles (EVs), is beginning to be explored. This review will summarize the recent literature describing the effects of exercise on hematopoiesis in individuals with obesity and introduce the potential contribution of EVs to this process.
Collapse
|
11
|
Callanan MC, Plummer HA, Green TM, Opitz T, Broderick T, Rendos N, Anz AW. Blood Flow Restriction Using a Pneumatic Tourniquet Is Not Associated With a Cellular Systemic Response. Arthrosc Sports Med Rehabil 2022; 4:e877-e882. [PMID: 35747643 PMCID: PMC9210389 DOI: 10.1016/j.asmr.2021.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 12/23/2021] [Indexed: 11/20/2022] Open
Abstract
Purpose The purpose of this study was to determine the effects of blood flow restriction (BFR) using a pneumatic tourniquet on CD34+ cells, platelets, white blood cells, neutrophils, lymphocytes, lactate, and glucose compared with standard exercise. Methods Fifteen healthy volunteers (8 males and 7 females, 28.6 ± 3.6 years old) who were able to perform the exercise sessions on a VersaClimber participated. Participants were randomized to undergo an experimental (EXP) occluded testing session using the pneumatic tourniquets on all 4 extremities and a control (CON) session. The exercise protocol concluded after 9 minutes or when participants reached a rating of perceived exertion of 20. Blood draws were performed before testing and immediately after the exercise session. Blood analysis consisted of complete blood counts as well as flow cytometry to measure peripheral CD34+ counts as a marker for hematopoietic progenitor cells (HPCs). Results A significant increase from before to after exercise values was observed in both the EXP and CON groups with CD34+, WBC counts, platelets, and lymphocytes; however, no differences existed between EXP and CON groups for any variable. CD34+ increased in the EXP (3.1 ± 1.6 vs. 4.3 ± 1.8 cells · L–1; P < .001) and CON (3.3 ± 1.9 vs. 4.4 ± 1.4 cells · L–1; P < .001) sessions. White blood cells also significantly increased in both the EXP (7.8 ± 1.4 vs. 11.8 ± 2.5 K · L–1 K · L–1; P < .001) and CON (7.5 ± 1.8 vs. 11.3 ± 3.0 K · L–1; P < .001) sessions. Platelets also increased in both the EXP (258.6 ± 52.5 vs. 309.9 ± 52.7 K · L–1; P < .001) and CON (263.1 ± 44.7 vs. 316.1 ± 43.9 K · L–1; P < .001) sessions, and conversely, a significant decrease in the average neutrophil counts in the EXP (mean difference = –13.7%; P < .001) and CON (mean difference = –13.2%; P < .001) sessions was observed. Lymphocyte counts in the EXP (mean difference = 22.8%; P < .001) and CON (mean difference = 19.3%; P < .001) sessions increased significantly. Conclusions There were no significant differences in systemic cellular responses when undergoing aerobic-based exercise with and without a pneumatic tourniquet system. Level of Evidence 2, prospective comparative study.
Collapse
|
12
|
González LM, Ospina LN, Sperling LE, Chaparro O, Cucarián JD. Therapeutic Effects of Physical Exercise and the Mesenchymal Stem Cell Secretome by Modulating Neuroinflammatory Response in Multiple Sclerosis. Curr Stem Cell Res Ther 2021; 17:621-632. [PMID: 34886779 DOI: 10.2174/1574888x16666211209155333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/14/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis (MS) is a neurodegenerative, demyelinating, and chronic inflammatory disease characterized by central nervous system (CNS) lesions that lead to high levels of disability and severe physical and cognitive disturbances. Conventional therapies are not enough to control the neuroinflammatory process in MS and are not able to inhibit ongoing damage to the CNS. Thus, the secretome of mesenchymal stem cells (MSC-S) has been postulated as a potential therapy that could mitigate symptoms and disease progression. We considered that its combination with physical exercise (EX) could induce superior effects and increase the MSC-S effectiveness in this condition. Recent studies have revealed that both EX and MSC-S share similar mechanisms of action that mitigate auto-reactive T cell infiltration, regulate the local inflammatory response, modulate the proinflammatory profile of glial cells, and reduce neuronal damage. Clinical and experimental studies have reported that these treatments in an isolated way also improve myelination, regeneration, promote the release of neurotrophic factors, and increase the recruitment of endogenous stem cells. Together, these effects reduce disease progression and improve patient functionality. Despite these results, the combination of these methods has not yet been studied in MS. In this review, we focus on molecular elements and cellular responses induced by these treatments in a separate way, showing their beneficial effects in the control of symptoms and disease progression in MS, as well as indicating their contribution in clinical fields. In addition, we propose the combined use of EX and MSC-S as a strategy to boost their reparative and immunomodulatory effects in this condition, combining their benefits on synaptogenesis, neurogenesis, remyelination, and neuroinflammatory response. The findings here reported are based on the scientific evidence and our professional experience that will bring significant progress to regenerative medicine to deal with this condition.
Collapse
Affiliation(s)
- Lina María González
- Physiotherapy Program, School of Medicine and Health Sciences, Universidad del Rosario AK 24 #63c-69, Bogotá. Colombia
| | - Laura Natalia Ospina
- Physiotherapy Program, School of Medicine and Health Sciences, Universidad del Rosario AK 24 #63c-69, Bogotá. Colombia
| | - Laura Elena Sperling
- Faculty of Pharmacy & Fundamental Health Science Institute, Federal University of Rio Grande do Sul Rua Ramiro Barcelos, 2600-Prédio Anexo - Floresta, Porto Alegre. Brazil
| | - Orlando Chaparro
- Physiology Department, Faculty of Medicine, Universidad Nacional de Colombia Ak 30 #45-03, Bogotá. Colombia
| | - Jaison Daniel Cucarián
- Physiotherapy Program, School of Medicine and Health Sciences, Universidad del Rosario AK 24 #63c-69, Bogotá. Colombia
| |
Collapse
|
13
|
Root SH, Aguila HL. Novel population of human monocyte and osteoclast progenitors from pluripotent stem cells and peripheral blood. Blood Adv 2021; 5:4435-4446. [PMID: 34581760 PMCID: PMC8579260 DOI: 10.1182/bloodadvances.2021004552] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoclasts are multinuclear cells of monocytic lineage, with the ability to resorb bone. Studies in mouse have identified bone marrow clonal progenitors able to generate mature osteoclast cells (OCs) in vitro and in vivo. These osteoclast progenitors (OCPs) can also generate macrophages and dendritic cells. Interestingly, cells with equivalent potential can be detected in periphery. In humans, cells with OCP activity have been identified in bone marrow and periphery; however, their characterization has not been as extensive. We have developed reproducible methods to derive, from human pluripotent stem cells, a population containing monocyte progenitors able to generate functional OCs. Within this population, we have identified cells with monocyte and osteoclast progenitor activity based on CD11b and CD14 expression. A population double positive for CD11b and CD14 contains cells with expected osteoclastic potential. However, the double negative (DN) population, containing most of the hematopoietic progenitor activity, also presents a very high osteoclastic potential. These progenitor cells can also be differentiated to macrophage and dendritic cells. Further dissection within the DN population identified cells bearing the phenotype CD15-CD115+ as the population with highest monocytic progenitor and osteoclastic potential. When similar methodology was used to identify OCPs from human peripheral blood, we confirmed a published OCP population with the phenotype CD11b+CD14+. In addition, we identified a second population (CD14-CD11bloCD115+) with high monocytic progenitor activity that was also able to form osteoclast like cells, similar to the 2 populations identified from pluripotent stem cells.
Collapse
Affiliation(s)
- Sierra H. Root
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT
| | - Héctor L. Aguila
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT
| |
Collapse
|
14
|
Zhao Y, Suo Y, Yang Z, Hao Y, Li W, Su Y, Shi Y, Gao Y, Song L, Yin X, Shi H. Inspiration for the prevention and treatment of neuropsychiatric disorders: New insight from the bone-brain-axis. Brain Res Bull 2021; 177:263-272. [PMID: 34678443 DOI: 10.1016/j.brainresbull.2021.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 11/30/2022]
Abstract
Bone is the main supporting structure of the body and the main organ involved in body movement and calcium and phosphorus metabolism. Recent studies have shown that bone is also a potential new endocrine organ that participates in the physiological and pathophysiological processes of the cardiovascular, digestive, and endocrine systems through various bioactive cytokines secreted by bone cells and bone marrow. Bone-derived active cytokines can also directly act on the central nervous system and regulate brain function and individual behavior. The bidirectional regulation of the bone-brain axis has gradually attracted attention in the field of neuroscience. This paper reviews the regulatory effects of bone-derived active cytokines and bone-derived cells on individual brain function and brain diseases, as well as the occurrence and development of related neuropsychiatric diseases. The central regulatory mechanism function is briefly introduced, which will broaden the scope for mechanistic research and help establish prevention and treatment strategies for neuropsychiatric diseases based on the bone-brain axis.
Collapse
Affiliation(s)
- Ye Zhao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yining Suo
- Child Health Department, Hebei Children's Hospital, Shijiazhuang 050031, China
| | - Zhenbang Yang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Ying Hao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Wenshuya Li
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yujiao Su
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Yuan Gao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Li Song
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Xi Yin
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Department of Functional Region of Diagnosis, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China.
| |
Collapse
|
15
|
Taylor GS, Shaw A, Smith K, Capper TE, Scragg JH, Cronin M, Bashir A, Flatt A, Campbell MD, Stevenson EJ, Shaw JA, Ross M, West DJ. Type 1 diabetes patients increase CXCR4 + and CXCR7 + haematopoietic and endothelial progenitor cells with exercise, but the response is attenuated. Sci Rep 2021; 11:14502. [PMID: 34267242 PMCID: PMC8282661 DOI: 10.1038/s41598-021-93886-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/25/2021] [Indexed: 01/01/2023] Open
Abstract
Exercise mobilizes angiogenic cells, which stimulate vascular repair. However, limited research suggests exercise-induced increase of endothelial progenitor cell (EPCs) is completely lacking in type 1 diabetes (T1D). Clarification, along with investigating how T1D influences exercise-induced increases of other angiogenic cells (hematopoietic progenitor cells; HPCs) and cell surface expression of chemokine receptor 4 (CXCR4) and 7 (CXCR7), is needed. Thirty T1D patients and 30 matched non-diabetes controls completed 45 min of incline walking. Circulating HPCs (CD34+, CD34+CD45dim) and EPCs (CD34+VEGFR2+, CD34+CD45dimVEGFR2+), and subsequent expression of CXCR4 and CXCR7, were enumerated by flow cytometry at rest and post-exercise. Counts of HPCs, EPCs and expression of CXCR4 and CXCR7 were significantly lower at rest in the T1D group. In both groups, exercise increased circulating angiogenic cells. However, increases was largely attenuated in the T1D group, up to 55% lower, with CD34+ (331 ± 437 Δcells/mL vs. 734 ± 876 Δcells/mL p = 0.048), CD34+VEGFR2+ (171 ± 342 Δcells/mL vs. 303 ± 267 Δcells/mL, p = 0.006) and CD34+VEGFR2+CXCR4+ (126 ± 242 Δcells/mL vs. 218 ± 217 Δcells/mL, p = 0.040) significantly lower. Exercise-induced increases of angiogenic cells is possible in T1D patients, albeit attenuated compared to controls. Decreased mobilization likely results in reduced migration to, and repair of, vascular damage, potentially limiting the cardiovascular benefits of exercise.Trial registration: ISRCTN63739203.
Collapse
Affiliation(s)
- Guy S Taylor
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andy Shaw
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kieran Smith
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tess E Capper
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Jadine H Scragg
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Michael Cronin
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Ayat Bashir
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Anneliese Flatt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew D Campbell
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, UK.,Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Emma J Stevenson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James A Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, UK
| | - Daniel J West
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
16
|
de Sire A, Marotta N, Marinaro C, Curci C, Invernizzi M, Ammendolia A. Role of Physical Exercise and Nutraceuticals in Modulating Molecular Pathways of Osteoarthritis. Int J Mol Sci 2021; 22:5722. [PMID: 34072015 PMCID: PMC8198532 DOI: 10.3390/ijms22115722] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling disease that affects millions of patients. Its etiology is largely unknown, but it is most likely multifactorial. OA pathogenesis involves the catabolism of the cartilage extracellular matrix and is supported by inflammatory and oxidative signaling pathways and marked epigenetic changes. To delay OA progression, a wide range of exercise programs and naturally derived compounds have been suggested. This literature review aims to analyze the main signaling pathways and the evidence about the synergistic effects of these two interventions to counter OA. The converging nutrigenomic and physiogenomic intervention could slow down and reduce the complex pathological features of OA. This review provides a comprehensive picture of a possible signaling approach for targeting OA molecular pathways, initiation, and progression.
Collapse
Affiliation(s)
- Alessandro de Sire
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| | - Nicola Marotta
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| | - Cinzia Marinaro
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| | - Claudio Curci
- Physical Medicine and Rehabilitation Unit, Department of Neurosciences, ASST Carlo Poma, 46100 Mantova, Italy;
| | - Marco Invernizzi
- Physical Medicine and Rehabilitation, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy;
- Translational Medicine, Dipartimento Attività Integrate Ricerca e Innovazione (DAIRI), Azienda Ospedaliera S.S. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Antonio Ammendolia
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| |
Collapse
|
17
|
Callanan MC, Christensen KD, Plummer HA, Torres J, Anz AW. Elevation of Peripheral Blood CD34+ and Platelet Levels After Exercise With Cooling and Compression. Arthrosc Sports Med Rehabil 2021; 3:e399-e410. [PMID: 34027448 PMCID: PMC8129037 DOI: 10.1016/j.asmr.2020.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/09/2020] [Indexed: 11/11/2022] Open
Abstract
Purpose To analyze the cellular response and chemokine profiles following exercise using cooling and blood flow restriction on the Vasper system. Methods Healthy male patients between the ages of 20 and 39 years were recruited. Testing was performed on the Vasper system, a NuStep cross-trainer with concomitant 4-limb venous compression with proximal arm cuffs at 40 mm Hg and proximal leg cuffs at 65 mm Hg. A cooling vest and cooling mat (8.3°C) were used. A 7-minute warm-up followed by alternating 30- and 60-second sprints with 1.5 and 2 minutes of active recovery, respectively, between each sprint. Peripheral blood was drawn before exercise, immediately following exercise (T20), 10 minutes after the first post-exercise blood draw (T30), and then every 30 minutes (T60, T90, T120, T150, T180). A blood draw occurred at 24 hours’ postexercise. Complete blood count, monoclonal flow cytometry for CD34+, and enzyme-linked immunosorbent assay were used to analyze the samples. Results Sixteen healthy male patients (29.5 ± 4.5years, 1.78 ± 0.05m, 83.7 ± 11.4 kg) were enrolled. There was an immediate, temporary increase in white blood cell counts, marked by an increase in lymphocyte differential (38.3 ± 6.5 to 44.3 ± 9.0%, P = .001), decrease in neutrophil differential (47.8 ± 6.6 to 42.0 ± 9.1%, P < .001), and platelets (239.5 ± 57.2 to 268.6 ± 86.3 K⋅μL–1, P = .01). Monocytes significantly decreased from PRE to T90 (9.8 ± 1.1 to 8.9 ± 1.1K/μL, P < .001) and T120 (8.9 ± 1.1 K/μL, P < .0001). There was a significant increase in CD34+ cells (3.9 ± 2.0 to 5.3 ± 2.8 cells⋅μL–1, P < .001). No detectable differences in measured cytokine levels of interleukin (IL)-10, IL-6, granulocyte-macrophage colony-stimulating factor , IL-1ra, tumor necrosis factor-α, or IL-2 were observed. Conclusions A significant elevation of peripheral blood CD34+ and platelet levels immediately following the exercise session was observed; however, there was no effect on peripheral circulation of IL-10, IL-6, IL-1ra, tumor necrosis factor-α, or IL-2. Clinical Relevance Exercise can be considered as a way to manipulate point-of-care blood products like platelet-rich plasma and may increase product yield.
Collapse
Affiliation(s)
| | | | | | | | - Adam W Anz
- Andrews Institute for Orthopedics & Sports Medicine, U.S.A
| |
Collapse
|
18
|
Schmid M, Kröpfl JM, Spengler CM. Changes in Circulating Stem and Progenitor Cell Numbers Following Acute Exercise in Healthy Human Subjects: a Systematic Review and Meta-analysis. Stem Cell Rev Rep 2021; 17:1091-1120. [PMID: 33389632 PMCID: PMC8316227 DOI: 10.1007/s12015-020-10105-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/22/2022]
Abstract
Despite of the increasing number of investigations on the effects of acute exercise on circulating stem and progenitor cell (SC) numbers, and in particular on respective subgroups, i.e. endothelial (ESC), hematopoietic (HSC), and mesenchymal (MSC) stem and progenitor cells, a consensus regarding mechanisms and extent of these effects is still missing. The aim of this meta-analysis was to systematically evaluate the overall-effects of acute exercise on the different SC-subgroups and investigate possible subject- and intervention-dependent factors affecting the extent of SC-mobilization in healthy humans. Trials assessing SC numbers before and at least one timepoint after acute exercise, were identified in a systematic computerized search. Compared to baseline, numbers were significantly increased for early and non-specified SCs (enSCs) until up to 0.5 h after exercise (0–5 min: +0.64 [Standardized difference in means], p < 0.001; 6–20 min: +0.42, p < 0.001; 0.5 h: +0.29, p = 0.049), for ESCs until 12–48 h after exercise (0–5 min: +0.66, p < 0.001; 6–20 min: +0.43 p < 0.001; 0.5 h: +0.43, p = 0.002; 1 h: +0.58, p = 0.001; 2 h: +0.50, p = 0.002; 3–8 h: +0.70, p < 0.001; 12–48 h: +0.38, p = 0.003) and for HSCs at 0–5 min (+ 0.47, p < 0.001) and at 3 h after exercise (+ 0.68, p < 0.001). Sex, intensity and duration of the intervention had generally no influence. The extent and kinetics of the exercise-induced mobilization of SCs differ between SC-subpopulations. However, also definitions of SC-subpopulations are non-uniform. Therefore, finding a consensus with a clear definition of cell surface markers defining ESCs, HSCs and MSCs is a first prerequisite for understanding this important topic. ![]()
Collapse
Affiliation(s)
- M Schmid
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - J M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - C M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
19
|
Callanan MC, Plummer HA, Chapman GL, Opitz TJ, Rendos NK, Anz AW. Blood Flow Restriction Training Using the Delfi System Is Associated With a Cellular Systemic Response. Arthrosc Sports Med Rehabil 2020; 3:e189-e198. [PMID: 33615264 PMCID: PMC7879193 DOI: 10.1016/j.asmr.2020.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 09/04/2020] [Indexed: 01/31/2023] Open
Abstract
Purpose To determine the effects of blood flow restriction (BFR) exercise on CD34+ cells, platelets, white blood cells, neutrophils, lymphocytes, lactate, and glucose. Methods Healthy participants aged 20 to 39 years who were able to perform the exercise sessions were recruited. Participants underwent an experimental (EXP) occluded testing session and a control (CON) session using the Delfi Personalized Tourniquet System. Blood draws were performed prior to testing and immediately after the exercise session. Blood analysis consisted of a complete blood count as well as flow cytometry to measure peripheral CD34+ counts as a marker for hematopoietic progenitor cells. Results Fourteen men (aged 30.8 ± 3.9 years) volunteered. There was a significant increase in average CD34+ counts immediately after the EXP session only (3.1 ± 1.2 cells ⋅ μL-1 vs 5.2 ± 2.9 cells ⋅ μL-1, P = .012). Platelet counts were significantly elevated after both sessions, with the average increase being higher after the EXP session (mean difference [MD], 34,200/μL; P < .002) than after the CON session (MD, 11,600/μL; P < .002). White blood cell counts significantly increased after both the EXP (8,400 ± 2,200/μL vs 6,300 ± 1,600/μL; P < .001) and CON (MD, 900/μL; P < .001) sessions. There was a significant increase from baseline to immediately after exercise in the average number of lymphocytes (MD, 6.3%; P < .001) and, conversely, a significant decrease in the average neutrophil count (MD, 6.5%; P < .001) in the EXP session only. Lactate levels significantly increased in the EXP (MD, 6.1 mmol ⋅ L-1; P = .001) and CON (MD, 3.6 mmol ⋅ L-1; P = .001) groups. No changes in glucose levels were observed. Conclusions Exercise with BFR causes a significant post-exercise increase in peripheral hematopoietic progenitor cells and platelets, beyond that of standard resistance training. Clinical Relevance BFR can be considered a way to manipulate point-of-care blood products such as platelet-rich plasma to increase product yield.
Collapse
Affiliation(s)
| | - Hillary A Plummer
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A
| | | | - Tyler J Opitz
- Andrews Institute for Orthopedics & Sports Medicine, Gulf Breeze, Florida, U.S.A
| | - Nicole K Rendos
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A
| | - Adam W Anz
- Andrews Institute for Orthopedics & Sports Medicine, Gulf Breeze, Florida, U.S.A
| |
Collapse
|
20
|
Smith JK. Exercise as an Adjuvant to Cartilage Regeneration Therapy. Int J Mol Sci 2020; 21:ijms21249471. [PMID: 33322825 PMCID: PMC7763351 DOI: 10.3390/ijms21249471] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
This article provides a brief review of the pathophysiology of osteoarthritis and the ontogeny of chondrocytes and details how physical exercise improves the health of osteoarthritic joints and enhances the potential of autologous chondrocyte implants, matrix-induced autologous chondrocyte implants, and mesenchymal stem cell implants for the successful treatment of damaged articular cartilage and subchondral bone. In response to exercise, articular chondrocytes increase their production of glycosaminoglycans, bone morphogenic proteins, and anti-inflammatory cytokines and decrease their production of proinflammatory cytokines and matrix-degrading metalloproteinases. These changes are associated with improvements in cartilage organization and reductions in cartilage degeneration. Studies in humans indicate that exercise enhances joint recruitment of bone marrow-derived mesenchymal stem cells and upregulates their expression of osteogenic and chondrogenic genes, osteogenic microRNAs, and osteogenic growth factors. Rodent experiments demonstrate that exercise enhances the osteogenic potential of bone marrow-derived mesenchymal stem cells while diminishing their adipogenic potential, and that exercise done after stem cell implantation may benefit stem cell transplant viability. Physical exercise also exerts a beneficial effect on the skeletal system by decreasing immune cell production of osteoclastogenic cytokines interleukin-1β, tumor necrosis factor-α, and interferon-γ, while increasing their production of antiosteoclastogenic cytokines interleukin-10 and transforming growth factor-β. In conclusion, physical exercise done both by bone marrow-derived mesenchymal stem cell donors and recipients and by autologous chondrocyte donor recipients may improve the outcome of osteochondral regeneration therapy and improve skeletal health by downregulating osteoclastogenic cytokine production and upregulating antiosteoclastogenic cytokine production by circulating immune cells.
Collapse
Affiliation(s)
- John Kelly Smith
- Departments of Academic Affairs and Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, P.O. Box 70300, Johnson City, TN 37614, USA
| |
Collapse
|
21
|
Exercise-Induced Myokines can Explain the Importance of Physical Activity in the Elderly: An Overview. Healthcare (Basel) 2020; 8:healthcare8040378. [PMID: 33019579 PMCID: PMC7712334 DOI: 10.3390/healthcare8040378] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022] Open
Abstract
Physical activity has been found to aid the maintenance of health in the elderly. Exercise-induced skeletal muscle contractions lead to the production and secretion of many small proteins and proteoglycan peptides called myokines. Thus, studies on myokines are necessary for ensuring the maintenance of skeletal muscle health in the elderly. This review summarizes 13 myokines regulated by physical activity that are affected by aging and aims to understand their potential roles in metabolic diseases. We categorized myokines into two groups based on regulation by aerobic and anaerobic exercise. With aging, the secretion of apelin, β-aminoisobutyric acid (BAIBA), bone morphogenetic protein 7 (BMP-7), decorin, insulin-like growth factor 1 (IGF-1), interleukin-15 (IL-15), irisin, stromal cell-derived factor 1 (SDF-1), sestrin, secreted protein acidic rich in cysteine (SPARC), and vascular endothelial growth factor A (VEGF-A) decreased, while that of IL-6 and myostatin increased. Aerobic exercise upregulates apelin, BAIBA, IL-15, IL-6, irisin, SDF-1, sestrin, SPARC, and VEGF-A expression, while anaerobic exercise upregulates BMP-7, decorin, IGF-1, IL-15, IL-6, irisin, and VEGF-A expression. Myostatin is downregulated by both aerobic and anaerobic exercise. This review provides a rationale for developing exercise programs or interventions that maintain a balance between aerobic and anaerobic exercise in the elderly.
Collapse
|
22
|
Schauer T, Hojman P, Gehl J, Christensen JF. Exercise training as prophylactic strategy in the management of neutropenia during chemotherapy. Br J Pharmacol 2020; 179:2925-2937. [DOI: 10.1111/bph.15141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/28/2020] [Accepted: 05/11/2020] [Indexed: 12/23/2022] Open
Affiliation(s)
- Tim Schauer
- Centre for Physical Activity Research, RigshospitaletUniversity of Copenhagen Copenhagen Denmark
| | - Pernille Hojman
- Centre for Physical Activity Research, RigshospitaletUniversity of Copenhagen Copenhagen Denmark
| | - Julie Gehl
- Center for Experimental Drug and Gene Electrotransfer (C*EDGE), Department of Clinical Oncology and Palliative CareZealand University Hospital Køge Denmark
- Faculty of Health and Medical Sciences, Department of Clinical MedicineUniversity of Copenhagen Copenhagen Denmark
| | - Jesper Frank Christensen
- Centre for Physical Activity Research, RigshospitaletUniversity of Copenhagen Copenhagen Denmark
| |
Collapse
|
23
|
Ewbank AC, Sacristán C, Costa-Silva S, Antonelli M, Lorenço JR, Nogueira GA, Ebert MB, Kolesnikovas CKM, Catão-Dias JL. Postmortem findings in Magellanic penguins (Spheniscus magellanicus) caught in a drift gillnet. BMC Vet Res 2020; 16:153. [PMID: 32448250 PMCID: PMC7245875 DOI: 10.1186/s12917-020-02363-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 05/10/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Penguin interaction with gillnets has been extensively reported in the Atlantic and Pacific Oceans, and is considered a major conservation threat. Among penguin species, Magellanic penguins (Spheniscus magellanicus) are currently considered of great concern, particularly in Brazil, where they are highly susceptible to gillnet bycatch. Nevertheless, information about drowning-associated microscopic findings in penguins is limited. RESULTS We describe the anatomopathological findings of 20 Magellanic penguins that drowned after getting entangled in a drift gillnet while wintering along the Brazilian shelf and washed ashore still enmeshed in Santa Catarina, Brazil. All 20 birds (19 juveniles and 1 adult; 18 females and 2 males) were in good body condition. Major gross findings were abrasion, bruising, and local erythema and edema of the wings, multiorgan congestion, jugular vein engorgement, pulmonary edema and hemorrhage, splenomegaly and hepatomegaly, fluid in the trachea, serous bloody fluid in the lungs, gastrointestinal parasites (nematodes, cestodes and trematodes), and debris in the stomach. The most common histopathological findings were cerebral and pulmonary congestion, pulmonary edema, splenic histiocytosis, lymphoid splenic hyperplasia, acute splenitis, extramedullary hepatic hematopoiesis, and parasitic enteritis. Although unspecific, the observed multiorgan congestion and pulmonary edema are consistent with previous reports of drowning in birds and may be indicative of this process. CONCLUSIONS Drowning may be a challenging diagnosis (e.g., carcass decomposition, predation), but must be considered as a differential in all beach-cast seabird postmortem examinations. To the authors' knowledge this is the largest anatomopathological study based on microscopic examination in drowned penguins.
Collapse
Affiliation(s)
- Ana Carolina Ewbank
- Laboratory of Wildlife Comparative Pathology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, SP, 05508-270, Brazil.
| | - Carlos Sacristán
- Laboratory of Wildlife Comparative Pathology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, SP, 05508-270, Brazil
| | | | | | | | | | - Mariana B Ebert
- Laboratory of Wildlife Parasitology (LAPAS), Parasitology Department, Biosciences Institute, São Paulo State University (UNESP), Botucatu, SP, 18618-000, Brazil
| | | | - José Luiz Catão-Dias
- Laboratory of Wildlife Comparative Pathology, Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, SP, 05508-270, Brazil
| |
Collapse
|
24
|
Kröpfl JM, Beltrami FG, Gruber HJ, Stelzer I, Spengler CM. Exercise-Induced Circulating Hematopoietic Stem and Progenitor Cells in Well-Trained Subjects. Front Physiol 2020; 11:308. [PMID: 32457637 PMCID: PMC7220991 DOI: 10.3389/fphys.2020.00308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 03/19/2020] [Indexed: 12/18/2022] Open
Abstract
It has been proposed that exercise-induced systemic oxidative stress increases circulating hematopoietic stem and progenitor cell (HPC) number in active participants, while HPC clonogenicity is reduced post-exercise. However, HPCs could be protected against exercise-induced reactive oxygen species in a trained state. Therefore, we characterized the acute exercise-induced HPC profile of well-trained participants including cell number, clonogenicity, and clearance. Twenty-one healthy, well-trained participants-12 runners, 9 cyclists; age 30.0 (4.3) years-performed a strenuous acute exercise session consisting of 4 bouts of 4-min high-intensity with 3-min low-intensity in-between, which is known to elicit oxidative stress. Average power/speed of intense phases was 85% of the peak achieved in a previous incremental test. Before and 10 min after exercise, CD34+/45dim cell number and clonogenicity, total oxidative (TOC), and antioxidative (TAC) capacities, as well as CD31 expression on detected HPCs were investigated. TOC significantly decreased from 0.093 (0.059) nmol/l to 0.083 (0.052) nmol/l post-exercise (p = 0.044). Although HPC proportions significantly declined below baseline (from 0.103 (0.037)% to 0.079 (0.028)% of mononuclear cells, p < 0.001), HPC concentrations increased post-exercise [2.10 (0.75) cells/μl to 2.46 (0.98) cells/μl, p = 0.002] without interaction between exercise modalities, while HPC clonogenicity was unaffected. Relating HPC concentrations and clonogenicity to exercise session specific (anti-) oxidative parameters, no association was found. CD31 median fluorescent intensity expression on detected HPCs was diminished post-exercise [from 1,675.9 (661.0) to 1,527.1 (558.9), p = 0.023] and positively correlated with TOC (r rm = 0.60, p = 0.005). These results suggest that acute exercise-reduced oxidative stress influences HPC clearance but not mobilization in well-trained participants. Furthermore, a well-trained state protected HPCs' clonogenicity from post-exercise decline.
Collapse
Affiliation(s)
- Julia M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Fernando G Beltrami
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Ingeborg Stelzer
- Institute of Medical and Chemical Laboratory Diagnostics, LKH Hochsteiermark, Leoben, Austria
| | - Christina M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
25
|
Collao N, Farup J, De Lisio M. Role of Metabolic Stress and Exercise in Regulating Fibro/Adipogenic Progenitors. Front Cell Dev Biol 2020; 8:9. [PMID: 32047748 PMCID: PMC6997132 DOI: 10.3389/fcell.2020.00009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Obesity is a major public health concern and is associated with decreased muscle quality (i.e., strength, metabolism). Muscle from obese adults is characterized by increases in fatty, fibrotic tissue that decreases the force producing capacity of muscle and impairs glucose disposal. Fibro/adipogenic progenitors (FAPs) are muscle resident, multipotent stromal cells that are responsible for muscle fibro/fatty tissue accumulation. Additionally, they are indirectly involved in muscle adaptation through their promotion of myogenic (muscle-forming) satellite cell proliferation and differentiation. In conditions similar to obesity that are characterized by chronic muscle degeneration, FAP dysfunction has been shown to be responsible for increased fibro/fatty tissue accumulation in skeletal muscle, and impaired satellite cell function. The role of metabolic stress in regulating FAP differentiation and paracrine function in skeletal muscle is just beginning to be unraveled. Thus, the present review aims to summarize the recent literature on the role of metabolic stress in regulating FAP differentiation and paracrine function in skeletal muscle, and the mechanisms responsible for these effects. Furthermore, we will review the role of physical activity in reversing or ameliorating the detrimental effects of obesity on FAP function.
Collapse
Affiliation(s)
- Nicolas Collao
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Michael De Lisio
- School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
26
|
Hematopoietic Stem and Progenitor Cell (HSPC) Mobilization Responses to Different Exercise Intensities in Young and Older Adults. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42978-019-00050-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
27
|
Cooper DM, Radom-Aizik S. Exercise-associated prevention of adult cardiovascular disease in children and adolescents: monocytes, molecular mechanisms, and a call for discovery. Pediatr Res 2020; 87:309-318. [PMID: 31649340 PMCID: PMC11177628 DOI: 10.1038/s41390-019-0581-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/07/2019] [Accepted: 08/15/2019] [Indexed: 12/28/2022]
Abstract
Atherosclerosis originates in childhood and adolescence. The goal of this review is to highlight how exercise and physical activity during childhood and adolescence, critical periods of growth and development, can prevent adult cardiovascular disease (CVD), particularly through molecular mechanisms of monocytes, a key cell of the innate immune system. Monocytes are heterogeneous and pluripotential cells that can, paradoxically, play a role in both the instigation and prevention of atherosclerosis. Recent discoveries in young adults reveal that brief exercise affects monocyte gene pathways promoting a cell phenotype that patrols the vascular system and repairs injuries. Concurrently, exercise inhibits pro-inflammatory monocytes, cells that contribute to vascular damage and plaque formation. Because CVD is typically asymptomatic in youth, minimally invasive techniques must be honed to study the subtle anatomic and physiologic evidence of vascular dysfunction. Exercise gas exchange and heart rate measures can be combined with ultrasound assessments of vascular anatomy and reactivity, and near-infrared spectroscopy to quantify impaired O2 transport that is often hidden at rest. Combined with functional, transcriptomic, and epigenetic monocyte expression and measures of monocyte-endothelium interaction, molecular mechanisms of early CVD can be formulated, and then translated into effective physical activity-based strategies in youth to prevent adult-onset CVD.
Collapse
Affiliation(s)
- Dan M Cooper
- Pediatric Exercise and Genomics Research Center, University of California Irvine School of Medicine, Pediatrics, Irvine, CA, USA.
| | - Shlomit Radom-Aizik
- Pediatric Exercise and Genomics Research Center, University of California Irvine School of Medicine, Pediatrics, Irvine, CA, USA
| |
Collapse
|
28
|
Abstract
Enforced egress of hematopoietic stem cells (HSCs) out of the bone marrow (BM) into the peripheral circulation, termed mobilization, has come a long way since its discovery over four decades ago. Mobilization research continues to be driven by the need to optimize the regimen currently available in the clinic with regard to pharmacokinetic and pharmacodynamic profile, costs, and donor convenience. In this review, we describe the most recent findings in the field and how we anticipate them to affect the development of mobilization strategies in the future. Furthermore, the significance of mobilization beyond HSC collection, i.e. for chemosensitization, conditioning, and gene therapy as well as a means to study the interactions between HSCs and their BM microenvironment, is reviewed. Open questions, controversies, and the potential impact of recent technical progress on mobilization research are also highlighted.
Collapse
Affiliation(s)
- Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine,, St. Louis, Missouri, 63110, USA
| |
Collapse
|
29
|
Emmons R, Ngu M, Xu G, Hernández-Saavedra D, Chen H, DE Lisio M. Effects of Obesity and Exercise on Bone Marrow Progenitor Cells after Radiation. Med Sci Sports Exerc 2019; 51:1126-1136. [PMID: 30640286 DOI: 10.1249/mss.0000000000001894] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The late effects of radiation therapy can have significant consequences for the health and quality of life of long-term cancer survivors. Radiation induces persistent alterations in hematopoietic stem and progenitor cells (HSPC) and the bone marrow environment; however, how relevant host factors such as obesity and exercise differentially regulate HSPC content and the bone marrow environment after radiation exposure remains unknown. The purpose of this investigation was to evaluate how the combination of obesity and exercise training modulates HSPC and their niche after sublethal radiation exposure in mice. METHODS Mice fed either a control or a high-fat diet to induce obesity remained sedentary or underwent a progressive treadmill exercise program. At 13 wk of age, mice were irradiated (3 Gy) and continued their specific diets and exercise program for four more weeks. RESULTS Exercise-trained mice had significantly higher quantities of several HSPC subpopulations and bone marrow stromal cell populations, whereas HSPC subpopulations were significantly lower in obese mice after radiation. Reactive oxygen species content was significantly decreased in HSPC with exercise training. Proteomics analysis of bone marrow supernatant revealed clustering of biologically relevant changes in exercise-trained mice. Functional evaluation of bone marrow supernatant revealed a significant increase in leukemia blast viability in obese mice but not in the exercise-trained mice (P < 0.05). CONCLUSION Together, these data suggest that exercise training partially restores the negative effects of obesity on HSPC and their niche after radiation exposure. As such, exercise training should be considered to mitigate the late effects of radiation therapy on the hematopoietic system for cancer survivors with or without obesity who have undergone radiation therapy.
Collapse
Affiliation(s)
- Russell Emmons
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Matthew Ngu
- School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA
| | - Guanying Xu
- Department of Food Sciences and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL
| | | | - Hong Chen
- Department of Food Sciences and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Michael DE Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL.,School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA.,Regenerative Medicine Program, Centre on Neuromuscular Disease, and Brain and Mind Institute, University of Ottawa, Ottawa, ON, CANADA
| |
Collapse
|
30
|
Sen S. Adult Stem Cells: Beyond Regenerative Tool, More as a Bio-Marker in Obesity and Diabetes. Diabetes Metab J 2019; 43:744-751. [PMID: 31902144 PMCID: PMC6943270 DOI: 10.4093/dmj.2019.0175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/17/2019] [Indexed: 12/23/2022] Open
Abstract
Obesity, diabetes, and cardiovascular diseases are increasing rapidly worldwide and it is therefore important to know the effect of exercise and medications for diabetes and obesity on adult stem cells. Adult stem cells play a major role in remodeling and tissue regeneration. In this review we will focus mainly on two adult stem/progenitor cells such as endothelial progenitor cells and mesenchymal stromal cells in relation to aerobic exercise and diabetes medications, both of which can alter the course of regeneration and tissue remodelling. These two adult precursor and stem cells are easily obtained from peripheral blood or adipose tissue depots, as the case may be and are precursors to endothelium and mesenchymal tissue (fat, bone, muscle, and cartilage). They both are key players in maintenance of cardiovascular and metabolic homeostasis and can act also as useful biomarkers.
Collapse
Affiliation(s)
- Sabyasachi Sen
- Division of Endocrinology, Department of Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
31
|
Circulating adult stem and progenitor cell numbers-can results be trusted? Stem Cell Res Ther 2019; 10:305. [PMID: 31623690 PMCID: PMC6798345 DOI: 10.1186/s13287-019-1403-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/16/2019] [Accepted: 09/02/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Within the last years, the interest in physical exercise as non-invasive stimulus influencing circulating hematopoietic stem and progenitor cell (CPC) concentrations has constantly grown. Cell estimates are often derived by determining the subgroup of CPC as percent lymphocytes (LYM) or mononuclear cells (MNC) via flow cytometry and back calculation over whole blood (WB) cell counts. However, results might depend on the used cell isolation technique and/or gating strategy. We aimed to investigate MNC loss and apoptosis during the flow cytometry sample preparation process preceded by either density gradient centrifugation (DGC) or red blood cell lysis (RBCL) and the potential difference between results derived from back calculation at different stages of cell isolation and from WB. METHODS Human blood was subjected to DGC and RBCL. Samples were stained for flow cytometry analysis of CPC (CD34+/CD45dim) and apoptosis analysis (Annexin V) of MNC and CPC subsets. MNC and LYM gating strategies were compared. RESULTS Both DGC as well as RBCL yielded comparable CPC concentrations independent of the gating strategy when back calculated over WB values. However, cell loss and apoptosis differed between techniques, where after DGC LYM, and monocyte (MONO) concentrations significantly decreased (p < 0.01 and p < 0.05, respectively), while after RBCL LYM concentrations significantly decreased (p < 0.05) and MONO concentrations increased (p < 0.001). LYM apoptosis was comparable between techniques, but MONO apoptosis was higher after DGC than RBCL (p < 0.001). CONCLUSIONS Investigated MNC counts (LYM/MONO ratio) after cell isolation and staining did not always mimic WB conditions. Thus, final CPC results should be corrected accordingly, especially when reporting live CPC concentrations after DGC; otherwise, the CPC regenerative potential in circulation could be biased. This is of high importance in the context of non-invasively induced CPC mobilization such as by acute physical exercise, since these cell changes are small and conclusions drawn from published results might affect further applications of physical exercise as non-invasive therapy.
Collapse
|
32
|
Increased Gene Expression of RUNX2 and SOX9 in Mesenchymal Circulating Progenitors Is Associated with Autophagy during Physical Activity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8426259. [PMID: 31737174 PMCID: PMC6815530 DOI: 10.1155/2019/8426259] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/22/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Lack of physical exercise is considered an important risk factor for chronic diseases. On the contrary, physical exercise reduces the morbidity rates of obesity, diabetes, bone disease, and hypertension. In order to gain novel molecular and cellular clues, we analyzed the effects of physical exercise on differentiation of mesenchymal circulating progenitor cells (M-CPCs) obtained from runners. We also investigated autophagy and telomerase-related gene expression to evaluate the involvement of specific cellular functions in the differentiation process. We performed cellular and molecular analyses in M-CPCs, obtained by a depletion method, of 22 subjects before (PRE RUN) and after (POST RUN) a half marathon performance. In order to prove our findings, we performed also in vitro analyses by testing the effects of runners' sera on a human bone marrow-derived mesenchymal stem (hBM-MSC) cell line. PCR array analyses of PRE RUN versus POST RUN M-CPC total RNAs put in evidence several genes which appeared to be modulated by physical activity. Our results showed that physical exercise promotes differentiation. Osteogenesis-related genes as RUNX2, MSX1, and SPP1 appeared to be upregulated after the run; data showed also increased levels of BMP2 and BMP6 expressions. SOX9, COL2A1, and COMP gene enhanced expression suggested the induction of chondrocytic differentiation as well. The expression of telomerase-associated genes and of two autophagy-related genes, ATG3 and ULK1, was also affected and correlated positively with MSC differentiation. These data highlight an attractive cellular scenario, outlining the role of autophagic response to physical exercise and suggesting new insights into the benefits of physical exercise in counteracting chronic degenerative conditions.
Collapse
|
33
|
Bourzac C, Bensidhoum M, Pallu S, Portier H. Use of adult mesenchymal stromal cells in tissue repair: impact of physical exercise. Am J Physiol Cell Physiol 2019; 317:C642-C654. [PMID: 31241985 PMCID: PMC6850997 DOI: 10.1152/ajpcell.00530.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
Physical exercise (PE) has unquestionable beneficial effects on health, which likely extend into several organ-to-cell physiological processes. At the cell scale, endogenous mesenchymal stromal cells (MSCs) contribute to tissue repair, although their repair capacities may be insufficient in paucicellular or severely damaged tissues. For this reason, MSC transplantation holds great promise for tissue repair. With the goals of understanding if PE has beneficial effects on MSC biology and if PE potentiates their role in tissue repair, we reviewed literature reports regarding the effects of PE on MSC properties (specifically, proliferation, differentiation, and homing) and of a combination of PE and MSC transplantation on tissue repair (specifically neural, cartilage, and muscular tissues). Contradictory results have been reported; interpretation is complicated because various and different species, cell sources, and experimental protocols, specifically exercise programs, have been used. On the basis of these data, the effects of exercise on MSC proliferation and differentiation depend on exercise characteristics (type, intensity, duration, etc.) and on the characteristics of the tissue from which the MSCs were collected. For the in vitro studies, the level of strain (and other details of the mechanical stimulus), the time elapsed between the end of exposure to strain and MSC collection, the age of the donors, as well as the passage number at which the MSCs are evaluated also play a role. The combination of PE and MSC engraftment improves neural, cartilage, and muscular tissue recovery, but it is not clear whether the effects of MSCs and exercise are additive or synergistic.
Collapse
Affiliation(s)
- Celine Bourzac
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Ecole Nationale Vétérinaire d'Alfort, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Maisons-Alfort, France
| | - Morad Bensidhoum
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
| | - Stephane Pallu
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Université d'Orléans, Le Collegium sciences et techniques (COST), Orléans, France
| | - Hugues Portier
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Université d'Orléans, Le Collegium sciences et techniques (COST), Orléans, France
| |
Collapse
|
34
|
Anz AW, Parsa RS, Romero-Creel MF, Nabors A, Tucker MS, Harrison RM, Matuska AM. Exercise-Mobilized Platelet-Rich Plasma: Short-Term Exercise Increases Stem Cell and Platelet Concentrations in Platelet-Rich Plasma. Arthroscopy 2019; 35:192-200. [PMID: 30611351 DOI: 10.1016/j.arthro.2018.06.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate the effects of vigorous short-term exercise on the platelet and other cellular components of 2 point-of-care blood-processing devices: a buffy coat-based platelet-rich plasma (PRP) product and a plasma-based PRP product. METHODS Twenty healthy subjects (aged 21-45 years) participated in a 20-minute vigorous exercise regimen on an upright stationary bike at 70% to 85% of maximum target heart rate. Pre- and post-exercise blood was processed in either a plasma-based or automated buffy coat-based PRP system. Complete blood counts were used to compare the cellular components in whole blood and the PRP products. RESULTS Exercise significantly increased the concentrations of platelets by over 20% in whole blood (P < .001) and in both PRP products (P = .002 and P = .018). Both devices performed consistently with pre- and post-exercise blood. Buffy coat-based PRP prepared after exercise was also significantly larger in volume and had a significantly higher concentration of mobilized hematopoietic stem cells (hematopoietic progenitor cells [HPCs], from 1.7/μL to 2.7/μL, P = .043). The concentrations of all white blood cell types were increased, which could be differentially collected in the devices studied. CONCLUSIONS Exercise can be used to consistently alter the composition of PRP. Twenty minutes of vigorous exercise can increase platelet concentrations in plasma-based and buffy coat-based PRP products and can increase HPC concentrations and volume in buffy coat-based PRP. CLINICAL RELEVANCE This study shows a nonpharmacologic method to increase platelet and HPC harvests from peripheral blood. This is important because it highlights a method for altering biological therapies with limited comorbidity.
Collapse
Affiliation(s)
- Adam W Anz
- Andrews Research and Education Foundation, Gulf Breeze, U.S.A..
| | - Ronna S Parsa
- Andrews Research and Education Foundation, Gulf Breeze, U.S.A
| | | | | | | | | | | |
Collapse
|
35
|
Spielmann G, Agha N, Kunz H, Simpson RJ, Crucian B, Mehta S, Laughlin M, Campbell J. B cell homeostasis is maintained during long-duration spaceflight. J Appl Physiol (1985) 2018; 126:469-476. [PMID: 30496712 DOI: 10.1152/japplphysiol.00789.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Long-duration spaceflights reportedly induce immune dysregulation, which is considered a risk to astronaut safety and mission success. Recent studies have examined the impact of spaceflight on markers of adaptive and innate immunity, but no study, to date, has comprehensively evaluated humoral immunity and serological markers of B cell function. The aim of this study was to characterize changes in B cell numbers and phenotypes, along with plasma Igs and polyclonal free light chains (FLCs)-near-"real-time" biomarkers of Ig synthesis-in response to an ~6-mo mission to the International Space Station (ISS). Whole-blood samples were collected before flight, during flight ("Early flight," "Mid-flight," and "Late flight"), immediately upon return, and during a recovery period (R + 18, R + 30/R + 33, and R + 60/R + 66) from 23 ISS crew members. B Cell counts and phenotypes were measured throughout the duration of the mission, along with total plasma Ig and FLC levels. There was no effect of spaceflight on the number and proportion of the different B cell subsets. There was no difference in kappa FLC between preflight samples and either in-flight or recovery samples ( P > 0.05), and only a marginal reduction was observed in lambda FLC levels upon return to Earth ( P < 0.05). Furthermore, IgG and IgM remained unchanged during and after spaceflight compared with preflight values ( P > 0.05). Of note, plasma IgA concentrations were elevated in-flight compared with baseline and recovery values ( P < 0.05). These results indicate that B cell homeostasis is maintained during long-duration spaceflight, advocating for potential in-flight vaccination as viable countermeasures against viral reactivation during exploration-class missions.
Collapse
Affiliation(s)
| | - Nadia Agha
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston , Houston, Texas
| | - Hawley Kunz
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston , Houston, Texas.,Division of Endocrinology and Metabolism, Mayo Clinic, College of Medicine , Rochester, Minnesota
| | - Richard J Simpson
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston , Houston, Texas.,Department of Nutritional Sciences, The University of Arizona , Tucson, Arizona.,Department of Pediatrics, The University of Arizona , Tucson, Arizona.,Department of Immunobiology, The University of Arizona , Tucson, Arizona
| | - Brian Crucian
- National Aeronautics and Space Administration-Johnson Space Center , Houston, Texas
| | - Satish Mehta
- National Aeronautics and Space Administration-Johnson Space Center , Houston, Texas
| | - Mitzi Laughlin
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston , Houston, Texas.,Fondren Orthopedic Group, Fondren Orthopedic Research Institute , Houston, Texas
| | - John Campbell
- School of Kinesiology, Louisiana State University , Baton Rouge, Louisiana.,Department for Health, University of Bath , Bath , United Kingdom
| |
Collapse
|
36
|
Heinonen I, Boushel R, Hellsten Y, Kalliokoski K. Regulation of bone blood flow in humans: The role of nitric oxide, prostaglandins, and adenosine. Scand J Med Sci Sports 2018; 28:1552-1558. [PMID: 29377406 DOI: 10.1111/sms.13064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2017] [Indexed: 12/23/2022]
Abstract
The mechanisms that regulate bone blood flow (BBF) in humans are largely unknown. Animal studies suggest that nitric oxide (NO) could be involved, and in this study, we investigated the effects of inhibition of nitric oxide synthase (NOS) alone and in combination with inhibition of cyclooxygenase (COX) enzyme, thus prostaglandin (PG) synthesis on femoral bone marrow blood flow by positron emission tomography in healthy young men at rest and during one-leg dynamic exercise. In an additional group of healthy men, the role of adenosine (ADO) in the regulation of BBF during exercise was investigated by use of an adenosine receptor blocker (aminophylline). Inhibitors were directly infused into the femoral artery. Resting BBF was 1.1 ± 0.4 mL 100 g-1 min-1 and increased to almost sixfold in response to exercise (6.3 ± 1.5 mL 100 g-1 min-1 ). Inhibition of NOS reduced BBF at rest to 0.7 ± 0.3 mL 100 g-1 min-1 (P = .036), but did not affect BBF significantly during exercise (5.5 ± 1.4 mL 100 g-1 min-1 , P = .25). On the other hand, while combined NOS and COX inhibition did not cause any further reduction of blood flow at rest (0.6 ± 0.2 mL 100 g-1 min-1 ), the combined blockade reduced BBF during exercise by ~21%, to 5.0 ± 1.8 mL 100 g-1 min-1 (P = .014). Finally, the ADO inhibition during exercise reduced BBF from 5.5 ± 1.9 mL 100 g-1 min-1 to 4.6 ± 1.2 mL 100 g-1 min-1 (P = .045). In conclusion, our results support the view that NO is involved in controlling bone marrow blood flow at rest, and NO, PG, and ADO play important roles in controlling human BBF during exercise.
Collapse
Affiliation(s)
- I Heinonen
- Turku PET Centre, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, University of Turku, Turku, Finland.,Division of Experimental Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - R Boushel
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Y Hellsten
- Exercise and Sport Sciences, Section of Human Physiology, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
37
|
Koelwyn GJ, Quail DF, Zhang X, White RM, Jones LW. Exercise-dependent regulation of the tumour microenvironment. Nat Rev Cancer 2017; 17:620-632. [PMID: 28943640 DOI: 10.1038/nrc.2017.78] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The integrity and composition of the tumour microenvironment (TME) is highly plastic, undergoing constant remodelling in response to instructive signals derived from alterations in the availability and nature of systemic host factors. This 'systemic milieu' is directly modulated by host exposure to modifiable lifestyle factors such as exercise. Host exposure to regular exercise markedly reduces the risk of the primary development of several cancers and might improve clinical outcomes following a diagnosis of a primary disease. However, the molecular mechanisms that underpin the apparent antitumour effects of exercise are poorly understood. In this Opinion article, we explore the putative effects of exercise in reprogramming the interaction between the host and the TME. Specifically, we speculate on the possible effects of exercise on reprogramming 'distant' tissue microenvironments (those not directly involved in the exercise response) by analysing how alterations in the systemic milieu might modulate key TME components to influence cancer hallmarks.
Collapse
Affiliation(s)
- Graeme J Koelwyn
- NYU Langone Medical Center, Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, 550 First Avenue, New York, New York 10016, USA
| | - Daniela F Quail
- Goodman Cancer Research Centre, McGill University; and at the Department of Physiology, McGill University, 1160 Pine Avenue West, Montreal, Quebec H3A 1A3, Canada
| | - Xiang Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine; and at the Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Lee W Jones
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA; and at the Weil Cornell Medical Center, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
38
|
Booth SA, Charchar FJ. Cardiac telomere length in heart development, function, and disease. Physiol Genomics 2017; 49:368-384. [DOI: 10.1152/physiolgenomics.00024.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Telomeres are repetitive nucleoprotein structures at chromosome ends, and a decrease in the number of these repeats, known as a reduction in telomere length (TL), triggers cellular senescence and apoptosis. Heart disease, the worldwide leading cause of death, often results from the loss of cardiac cells, which could be explained by decreases in TL. Due to the cell-specific regulation of TL, this review focuses on studies that have measured telomeres in heart cells and critically assesses the relationship between cardiac TL and heart function. There are several lines of evidence that have identified rapid changes in cardiac TL during the onset and progression of heart disease as well as at critical stages of development. There are also many factors, such as the loss of telomeric proteins, oxidative stress, and hypoxia, that decrease cardiac TL and heart function. In contrast, antioxidants, calorie restriction, and exercise can prevent both cardiac telomere attrition and the progression of heart disease. TL in the heart is also indicative of proliferative potential and could facilitate the identification of cells suitable for cardiac rejuvenation. Although these findings highlight the involvement of TL in heart function, there are important questions regarding the validity of animal models, as well as several confounding factors, that need to be considered when interpreting results and planning future research. With these in mind, elucidating the telomeric mechanisms involved in heart development and the transition to disease holds promise to prevent cardiac dysfunction and potentiate regeneration after injury.
Collapse
Affiliation(s)
- S. A. Booth
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
| | - F. J. Charchar
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
- Department of Physiology, The University of Melbourne, Melbourne, Australia; and
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
39
|
Inoue A, Cheng XW, Huang Z, Hu L, Kikuchi R, Jiang H, Piao L, Sasaki T, Itakura K, Wu H, Zhao G, Lei Y, Yang G, Zhu E, Li X, Sato K, Koike T, Kuzuya M. Exercise restores muscle stem cell mobilization, regenerative capacity and muscle metabolic alterations via adiponectin/AdipoR1 activation in SAMP10 mice. J Cachexia Sarcopenia Muscle 2017; 8:370-385. [PMID: 27897419 PMCID: PMC5476856 DOI: 10.1002/jcsm.12166] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/08/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Exercise train (ET) stimulates muscle response in pathological conditions, including aging. The molecular mechanisms by which exercise improves impaired adiponectin/adiponectin receptor 1 (AdipoR1)-related muscle actions associated with aging are poorly understood. Here we observed that in a senescence-accelerated mouse prone 10 (SAMP10) model, long-term ET modulated muscle-regenerative actions. METHODS 25-week-old male SAMP10 mice were randomly assigned to the control and the ET (45 min/time, 3/week) groups for 4 months. Mice that were maintained in a sedentary condition served controls. RESULTS ET ameliorated aging-related muscle changes in microstructure, mitochondria, and performance. The amounts of proteins or mRNAs for p-AMPKα, p-Akt, p-ERK1/2, p-mTOR, Bcl-XL, p-FoxO3, peroxisome proliferators-activated receptor-γ coactivator, adiponectin receptor1 (adpoR1), and cytochrome c oxidase-IV, and the numbers of CD34+ /integrin-α7+ muscle stem cells (MuSCs) and proliferating cells in the muscles and bone-marrow were enhanced by ET, whereas the levels of p-GSK-3α and gp91phox proteins and apoptotic cells were reduced by ET. The ET also resulted in increased levels of plasma adiponectin and the numbers of bone-marrow (BM)-derived circulating CD34+ /integrin-α7+ MuSCs and their functions. Integrin-α7+ MuSCs of exercised mice had improved changes of those beneficial molecules. These ET-mediated aged muscle benefits were diminished by adiponectin and AdipoR1 blocking as well as AMPK inhibition. Finally, recombinant mouse adiponectin enhanced AMPK and mTOR phosphorylations in BM-derived integrin-α7+ cells. CONCLUSIONS These findings suggest that ET can improve aging-related impairments of BM-derived MuSC regenerative capacity and muscle metabolic alterations via an AMPK-dependent mechanism that is mediated by an adiponectin/AdipoR1 axis in SAMP10 mice.
Collapse
Affiliation(s)
- Aiko Inoue
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Xian Wu Cheng
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China.,Department of Cardiovascular of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Fukuoka, Japan
| | - Lina Hu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Ryosuke Kikuchi
- Department of Medical Technique, Nagoya University Hospital, Nagoya, 466-8550, Aichiken, Japan
| | - Haiying Jiang
- Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji, 133000, Jilin PR., China
| | - Limei Piao
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Shizuokaken, Japan
| | - Kohji Itakura
- Division for Medical Research Engineering, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Hongxian Wu
- Department of Sport Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Guangxian Zhao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Yanna Lei
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Guang Yang
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Enbo Zhu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Xiang Li
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, 133000, Jilin PR., China
| | - Kohji Sato
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Shizuokaken, Japan
| | - Teruhiko Koike
- Department of Sport Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Aichiken, Japan
| |
Collapse
|
40
|
Niemiro GM, Parel J, Beals J, van Vliet S, Paluska SA, Moore DR, Burd NA, De Lisio M. Kinetics of circulating progenitor cell mobilization during submaximal exercise. J Appl Physiol (1985) 2017; 122:675-682. [DOI: 10.1152/japplphysiol.00936.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/23/2016] [Accepted: 01/08/2017] [Indexed: 12/28/2022] Open
Abstract
Circulating progenitor cells (CPCs) are a heterogeneous population of stem/progenitor cells in peripheral blood that includes hematopoietic stem and progenitor cells (HSPCs and HSCs), endothelial progenitor cells (EPCs), and mesenchymal stem cells (MSCs) that are involved in tissue repair and adaptation. CPC mobilization during exercise remains uncharacterized in young adults. The purpose of this study was to investigate the kinetics of CPC mobilization during and after submaximal treadmill running and their relationship to mobilization factors. Seven men [age = 25.3 ± 2.4 yr, body mass index = 23.5 ± 1.0 kg/m2, peak O2uptake (V̇o2peak) = 60.9 ± 2.74 ml·kg−1·min−1] ran on a treadmill for 60 min at 70% V̇o2peak. Blood sampling occurred before (Pre), during [20 min (20e), 40 min (40e), 60 min (60e)], and after exercise [15 min (15p), 60 min (60p), 120 min (120p)] for quantification of CPCs (CD34+), HSPCs (CD34+/CD45low), HSCs (CD34+/CD45low/CD38−), CD34+MSCs (CD45−/CD34+/CD31−/CD105+), CD34−MSCs (CD45−/CD34−/CD31−/CD105+), and EPCs (CD45−/CD34+/CD31+) via flow cytometry. CPC concentration increased compared with Pre at 20e and 40e (2.7- and 2.4-fold, respectively, P < 0.05). HSPCs and HSCs increased at 20e compared with 60p (2.7- and 2.8-fold, respectively, P < 0.05), whereas EPCs and both MSC populations did not change. CXC chemokine ligand (CXCL) 12 (1.5-fold; P < 0.05) and stem cell factor (1.3-fold; P < 0.05) were increased at 40e and remained elevated postexercise. The peak increase in CPCs was positively correlated to concentration of endothelial cells during exercise with no relationship to CXCL12 and SCF. Our data show the kinetics of progenitor cell mobilization during exercise that could provide insight into cellular mediators of exercise-induced adaptations, and have implication for the use of exercise as an adjuvant therapy for CPC collection in hematopoietic stem cell transplant.NEW & NOTEWORTHY Using a comprehensive evaluation of circulating progenitor cells (CPCs), we show that CPC mobilization during exercise is related to tissue damage, and not plasma concentrations of CXC chemokine ligand 12 and stem cell factor. These data have implications for the use of exercise interventions as adjuvant therapy for CPC mobilization in the context of hematopoietic stem cell transplant and also support the role of mobilized progenitor cells as cellular mediators of systemic adaptations to exercise.
Collapse
Affiliation(s)
- Grace M. Niemiro
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Justin Parel
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Joseph Beals
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Stephan van Vliet
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Scott A. Paluska
- Department of Family Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Daniel R. Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada; and
| | - Nicholas A. Burd
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
- School of Human Kinetics, Brain and Mind Institute, Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
41
|
Baker JM, Nederveen JP, Parise G. Aerobic exercise in humans mobilizes HSCs in an intensity-dependent manner. J Appl Physiol (1985) 2017; 122:182-190. [PMID: 27881669 PMCID: PMC5283849 DOI: 10.1152/japplphysiol.00696.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/01/2016] [Accepted: 11/18/2016] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem and progenitor cells are necessary to maintain, repair, and reconstitute the hematopoietic blood cell system. Mobilization of these cells from bone marrow to blood can be greatly increased under certain conditions, one such being exercise. The purpose of this study was to identify the importance of exercise intensity in hematopoietic mobilization, to better understand the mobilization kinetics postexercise, and to determine if exercise is capable of mobilizing several specific populations of hematopoietic cells that have clinical relevance in a transplant setting. Healthy individuals were exercised on a cycle ergometer at 70% of their peak work rate (WRpeak) until volitional fatigue and at 30% of their WRpeak work matched to the 70% WRpeak bout. Blood was collected before, immediately post, and 10, 30, and 60 min postexercise. Total blood cells, hematocrit, and mononuclear cells isolated by density gradient centrifugation were counted. Specific populations of hematopoietic stem cells were analyzed by flow cytometry. Mononuclear cells, CD34+, CD34+/CD38-, CD34+/CD110+, CD3-/CD16+/CD56+, CD11c+/CD123-, and CD11c-/CD123+ cells per millilter of blood increased postexercise. Overall, the 70% WRpeak exercise group showed greater mobilization immediately postexercise, while there was no observable increase in mobilization in the work matched 30% WRpeak exercise group. Mobilization of specific populations of hematopoietic cells mirrored changes in the general mobilization of mononuclear cells, suggesting that exercise serves as a nonspecific mobilization stimulus. Evidently, higher intensity exercise is capable of mobilizing hematopoietic cells to a large extent and immediately postexercise is an ideal time point for their collection. NEW & NOTEWORTHY Here we demonstrate for the first time that mobilization of hematopoietic stem cells (HSCs) through exercise is intensity dependent, with the greatest mobilization occurring immediately after high-intensity exercise. As well, we show that exercise is a general stimulus for mobilization: increases in specific HSC populations are reliant on general mononuclear cell mobilization. Finally, we demonstrate no differences in mobilization between groups with different aerobic fitness.
Collapse
Affiliation(s)
- Jeff M Baker
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
| | - Joshua P Nederveen
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
| | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada; and
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
42
|
Peake JM, Neubauer O, Walsh NP, Simpson RJ. Recovery of the immune system after exercise. J Appl Physiol (1985) 2016; 122:1077-1087. [PMID: 27909225 DOI: 10.1152/japplphysiol.00622.2016] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/31/2016] [Accepted: 11/16/2016] [Indexed: 12/27/2022] Open
Abstract
The notion that prolonged, intense exercise causes an "open window" of immunodepression during recovery after exercise is well accepted. Repeated exercise bouts or intensified training without sufficient recovery may increase the risk of illness. However, except for salivary IgA, clear and consistent markers of this immunodepression remain elusive. Exercise increases circulating neutrophil and monocyte counts and reduces circulating lymphocyte count during recovery. This lymphopenia results from preferential egress of lymphocyte subtypes with potent effector functions [e.g., natural killer (NK) cells, γδ T cells, and CD8+ T cells]. These lymphocytes most likely translocate to peripheral sites of potential antigen encounter (e.g., lungs and gut). This redeployment of effector lymphocytes is an integral part of the physiological stress response to exercise. Current knowledge about changes in immune function during recovery from exercise is derived from assessment at the cell population level of isolated cells ex vivo or in blood. This assessment can be biased by large changes in the distribution of immune cells between blood and peripheral tissues during and after exercise. Some evidence suggests that reduced immune cell function in vitro may coincide with changes in vivo and rates of illness after exercise, but more work is required to substantiate this notion. Among the various nutritional strategies and physical therapies that athletes use to recover from exercise, carbohydrate supplementation is the most effective for minimizing immune disturbances during exercise recovery. Sleep is an important aspect of recovery, but more research is needed to determine how sleep disruption influences the immune system of athletes.
Collapse
Affiliation(s)
- Jonathan M Peake
- School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia; .,Centre of Excellence for Applied Sport Science Research, Queensland Academy of Sport, Brisbane, Queensland, Australia
| | - Oliver Neubauer
- School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Neil P Walsh
- Extremes Research Group, School of Sport, Health and Exercise Sciences, Bangor University, Bangor, United Kingdom; and
| | - Richard J Simpson
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas
| |
Collapse
|
43
|
Puchert M, Adams V, Linke A, Engele J. Evidence for the involvement of the CXCL12 system in the adaptation of skeletal muscles to physical exercise. Cell Signal 2016; 28:1205-1215. [DOI: 10.1016/j.cellsig.2016.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 12/23/2022]
|
44
|
Exercise as an Adjuvant Therapy for Hematopoietic Stem Cell Mobilization. Stem Cells Int 2016; 2016:7131359. [PMID: 27123008 PMCID: PMC4830735 DOI: 10.1155/2016/7131359] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cell transplant (HSCT) using mobilized peripheral blood hematopoietic stem cells (HSPCs) is the only curative strategy for many patients suffering from hematological malignancies. HSPC collection protocols rely on pharmacological agents to mobilize HSPCs to peripheral blood. Limitations including variable donor responses and long dosing protocols merit further investigations into adjuvant therapies to enhance the efficiency of HSPCs collection. Exercise, a safe and feasible intervention in patients undergoing HSCT, has been previously shown to robustly stimulate HSPC mobilization from the bone marrow. Exercise-induced HSPC mobilization is transient limiting its current clinical potential. Thus, a deeper investigation of the mechanisms responsible for exercise-induced HSPC mobilization and the factors responsible for removal of HSPCs from circulation following exercise is warranted. The present review will describe current research on exercise and HSPC mobilization, outline the potential mechanisms responsible for exercise-induced HSPC mobilization, and highlight potential sites for HSPC homing following exercise. We also outline current barriers to the implementation of exercise as an adjuvant therapy for HSPC mobilization and suggest potential strategies to overcome these barriers.
Collapse
|