IMR Press / RCM / Volume 23 / Issue 12 / DOI: 10.31083/j.rcm2312396
Open Access Review
Circulating Endothelial Progenitor and Mesenchymal Stromal Cells as Biomarkers for Monitoring Disease Status and Responses to Exercise
Show Less
1 Research & Development, VA Medical Center, Washington, DC 20422, USA
2 Department of Health, Human Function, and Rehabilitation Sciences, The George Washington University, Washington, DC 20037, USA
3 Department of Medicine, VA Medical Center, Washington, DC 20422, USA
4 Department of Medicine, The George Washington University, Washington, DC 20037, USA
*Correspondence: ssen1@gwu.edu; sabyasachi.sen@va.gov (Sabyasachi Sen)
Academic Editor: Jerome L. Fleg
Rev. Cardiovasc. Med. 2022, 23(12), 396; https://doi.org/10.31083/j.rcm2312396
Submitted: 5 October 2022 | Revised: 13 November 2022 | Accepted: 15 November 2022 | Published: 2 December 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Noncommunicable chronic diseases, such as obesity, cardiovascular disease (CVD), and type 2 diabetes (T2D), pose significant health challenges globally. Important advances have been made in the understanding of the pathophysiologal mechanisms and treatment of noncommunicable diseases in recent years. Lack of physical activity is a primary contributor to many noncommunicable diseases including metabolic syndrome, T2D, CVD, and obesity. Certain diabetes medications and non-pharmaceutical interventions, such as physical activity and exercise, are shown to be effective in decreasing the CVD risks associated with heart disease, stroke, obesity, prediabetes, and T2D. The ability to measure and analyze circulating adult stem cells (ASCs) has gained particular interest due to their potential to identify at-risk individuals and implications in various therapeutics. Therefore, the purpose of this narrative review is to (1) provide an overview of ASCs; specifically endothelial progenitor cells (EPCs) and mesenchymal stromal cells (MSCs), (2) describe the responses of these cells to acute and chronic exercise, and (3) highlight the potential effect of exercise on EPCs and MSCs in aging and disease. EPCs are circulating cells, abundantly available in peripheral blood, bone marrow, and umbilical cord, and are defined by cell surface markers such as CD34+. EPCs are expected to play an important role in angiogenesis and neovascularization and have been implicated in the treatment of CVD. MSCs are essential for maintaining tissue and organ homeostasis. MSCs are defined as multipotent heterogeneous cells that can proliferate in vitro as plastic-adherent cells, have fibroblast-like morphology, form colonies in vitro, and can differentiate into ostyeoblasts, adipocytes, chondroblasts, and myoblasts. In the presence of aging and disease, EPCs and MSCs decrease in quantity and functional capacity. Importantly, exercise facilitates EPC differentiation and production from bone marrow and also helps to promote migration and homing to the hypoxic and damaged tissue which in turn improve angiogenesis and vasculogenesis. Similarly, exercise stimulates increases in proliferation and migratory activity of MSCs. Despite the reported benefits of exercise on EPC and MSC number and function, little is known regarding the optimal exercise prescription for aging and clinical populations. Moreover, the interactions between medications and exercise on EPCs and MSCs is currently unclear. Use of ASCs as a biomarker have the potential to revolutionize the management of patients with a variety of metabolic and obesity related disorders and also pro-inflammatory diseases. Further investigation of clinical entities are urgently needed to understand the implications of interventions such as exercise, diet, and various medications on EPC and MSC quantity and function in aging and clinical populations.

Keywords
endothelial progenitor cells
mesenchymal stromal cells
cardiorespiratory fitness
physical inactivity
resistance training
aerobic exercise
flow-mediated dilation
type 2 diabetes mellitus
prediabetes
obesity
1. Introduction

The World Health Organization (WHO) estimates nearly 450 billion people are suffering from diabetes globally [1]. As per the Centers for Disease Control and Prevention (CDC), nearly 96 million Americans have prediabetes with more than 37 million having diabetes [2]. Individuals with prediabetes are at high risk for developing type 2 diabetes (T2D), heart disease, and stroke [2]. The CDC also estimate that in the United States, 1 out of 3 adults suffer from high blood pressure, 1 out of every 20 deaths are due to stroke, and 1 out of every 4 deaths are due to coronary artery disease (CAD) [3, 4, 5]. Overweight and physical inactivity are two of the leading preventable risk factors for T2D. Being overweight or obese increases the risk for T2D, with 89.8% being overweight or having obesity (i.e., body mass index of 25 kg/m2 or higher) [2]. In addition, 34.3% of individuals diagnosed with T2D were classified as physically inactive, defined as getting less than 10 minutes a week of moderate or vigorous activity in each physical activity category of work, leisure time, and transportation [2]. The economic costs associated with T2D and obesity are substantial, being estimated at $327 billion for diagnosed T2D alone in the United States population [6]. These figures stress the importance of cardiometabolic complications associated with T2D and obesity and the potential implications on health care costs.

Lack of physical activity is a primary contributor to many noncommunicable diseases including metabolic syndrome, T2D, cardiovascular disease (CVD), and obesity [7, 8, 9, 10]. Robust evidence supports that high amounts of sedentary behavior increase the risk for all-cause and CVD mortality and T2D [11, 12, 13, 14, 15, 16]. For example, an inverse, non-linear dose-response relationship is observed between long-term leisure-time physical activity and all-cause and CVD mortality when assessed during up to 23-years of follow-up [14]. In adults, exercise capacity and energy expenditure are shown to be stronger predictors of all-cause mortality when compared to smoking, hypertension, obesity, and T2D [17]. Recently, Kokkinos et al. [15] showed that cardiorespiratory fitness is inversely associated with mortality in a graded fashion across age, sex, and race in a cohort of 750,302 United States Veterans during a median follow-up of 10.2 years. The lowest risk for mortality was seen at approximately 14 metabolic equivalents (METs) for men and women, with the risk for least fit individuals being 4-folder higher than the extremely fit individuals [15]. In addition to cardiorespiratory fitness, sufficient levels of muscular fitness is also found to have protective effects on all-cause and cancer mortality in healthy middle-aged men, men with hypertension, and patients with heart failure [18]. Importantly, it has been suggested that possessing higher muscular fitness may improve, to some extent, the adverse cardiovascular profile of overweight and obese individuals [18]. Despite the known benefits of engaging in physical activity and exercise, only 1 in 4 adults are estimated to meet the recommended levels of physical activity in the United States [19]. Among 383,928 adults (aged 18–80), only 23.5% were found to meet the physical activity guidelines of combined aerobic activity and muscle-strengthening activity [20]. Those individuals less likely to meet the physical activity guidelines tend to be older, women, current smokers, and have poorer self-rated health and lower education/income [20].

Certain diabetes medications and non-pharmaceutical interventions, such as physical activity and exercise, are shown to be effective in decreasing the CVD risks associated with heart disease, stroke, obesity, prediabetes, and T2D [10, 21, 22]. The United States based study Diabetes Prevention Program (DPP), lifestyle intervention significantly reduced the chances of progressing from prediabetes to T2D and risk of developing CVD [23]. According to the CDC, physical activity not only reduces the risk of developing overt T2D and CVD, but also helps to reduce body weight in individuals with obesity and has even been shown to reduce the risk of developing certain cancers associated with obesity [24]. Physical activity increases life expectancy irrespective of age, ethnicity, body shape or body mass [24, 25]. Compared to those who did not meet the physical activity guidelines, individuals who engaged in muscle-strengthening activities or aerobic activities were found to be at reduced risk of all cause mortality with larger survival benefits found in those engaged in both muscle-strengthening and aerobic activities [26]. Similar patterns were observed for cause specific mortality from CVD, cancer, and chronic lower respiratory tract diseases [26]. Importantly, evidence from a metaepidemiological study of 16 meta-analyses found exercise interventions to have similar benefits of many drug interventions for the secondary prevention of coronary heart disease, rehabilitation after stroke, treatment of heart failure, and prevention of T2D [27].

Currently, diabetes medications only evaluate the hypoglycemic effect of a medication and does not always evaluate its potential to improve endothelial function and regeneration. At best, only surrogates of endothelial function are used rather than evaluating cells of endothelial lineage or even hematopoietic lineage, pre- and post-medication. Historically, the U.S. Food and Drug Administration (FDA) did not require information regarding the effects of diabetes medications on cardiometabolic health although in the last 3–5 years that trend appears to be changing. Similarly, the standard practice for monitoring the effect of exercise in clinical practice is by assessing vascular structure and function using non-invasive measures (i.e., vessel size, blood pressure, flow-mediated dilation) or by analyzing plasma or serum biochemistry focusing on surrogates of endothelial function (i.e., interleukins, high-sensitivity C-reactive protein (hs-CRP), lipid profile). However, it may be more informative to directly study cells, such as adult stem cells (ASCs) of hematopoietic lineage, as these cells may serve as a valuable biomarker to detect and monitor the effect of disease status. Furthermore, the evaluation of ASCs in response to exercise may provide insight into the cellular mechanisms underlying the associated health benefits. Therefore, the purpose of this narrative review is to (1) provide an overview of ASCs; specifically endothelial progenitor cells (EPCs) and mesenchymal stromal cells (MSCs), (2) describe the responses of these cells to acute and chronic exercise, and (3) highlight the potential effects of exercise on EPCs and MSCs in aging and disease populations.

2. Adults Stem Cells

Stem cells are undifferentiated cells which can differentiate into specialized or specific cell type, tissue or organ. Predominantly, natural or non-genetically modified stem cells are divided into embryonic and somatic or ASCs. ASCs hold promise both as a regenerative tool, in a pro-apoptotic (and pro-inflammatory) state of prediabetes and T2D but also as a biomarker to monitor progression of a disease process from prediabetes to T2D, at a cellular level. For the purposes of this review, we will concentrate primarily on two types of ASCs, one is hematopoietic cells that may be precursor to mature EPCs and MSCs. Peripheral blood contains approximately 1% of mononuclear cells as EPCs. EPCs are circulating cells, abundantly available in peripheral blood, bone marrow, and umbilical cord. EPCs have been defined by cell surface markers such as CD34+ (a cell surface marker to delineate a cell that has progenitor-like capabilities). The cells that have dual marks such as CD34+ plus kinase domain receptor (KDR), or vascular endothelial growth factor-receptor-2 (VEGFR-2) have more markers that indicate a progenitor cell that has vascular cell-like properties. Other progenitor markers include CD133 and c-kit positivity. These cells are expected to have high mRNA gene expression for typical endothelial cells such as endothelial nitric oxide synthase (eNOS) and endothelial cell specific clotting factor agents such von-Willebrand’s factor (vWF). Therefore, EPCs that have mature endothelial cell-like properties of VEGFR-2, eNOS, and vWF positivity, are expected to play an important role in angiogenesis and neovascularization and have been implicated in the treatment of CVD [28, 29, 30].

MSCs are essential for maintaining tissue and organ homeostasis. MSCs are defined as multipotent heterogeneous cells that can proliferate in vitro as plastic-adherent cells, have fibroblast-like morphology, form colonies in vitro, and can differentiate into ostyeoblasts, adipocytes, chondroblasts, and myoblasts [31, 32, 33, 34, 35, 36]. Sources of obtaining MSC can vary from umbilical cord blood, bone marrow, adipose tissue, pancreatic islet, fetal liver and even the lung tissue [37, 38]. MSCs express CD105, CD73 and CD90 but not CD45, CD34, CD14, or CD11b, CD79a, or CD19 and HLA-DR surface molecules [36]. This is because the latter cell surface markers are thought to be of endothelial cell lineage. The mobilization and homing of MSCs (i.e., the migration and arrest within the vasculature of a tissue followed by transmigration across the endothelium and engraftment in the target tissue) is affected by systemic and inflammatory state [39]. Bone marrow and adipose tissue derived MSCs have been well established for MSC production for therapeutic purposes [31, 32, 34, 40]. For example, MSCs have been used in clinical trials for the treatment of various CVD’s such as ischemic heart disease, cerebro-vascular stroke, chronic kidney disease, and peripheral vascular disease (PAD) [34, 41]. MSCs are also involved in the processes that support skeletal muscle repair in response to resistance exercise [29, 42, 43].

3. EPCs and MSCs as Biomarkers of Aging and Disease

EPCs can act as a cellular biomarker that is more reliable than commonly used clinical serum-based markers for estimating and following endothelial dysfunction in aging, early T2D and prediabetes subjects, pre- and post-exercise, and even in response to medications [44, 45, 46, 47, 48, 49, 50, 51]. Older adults are shown to have lower baseline levels of HSC, EPC, angiogenic T cells, as well as chemokine receptor 4 expressing circulating angiogenic cells (CXCR4-expressing CACs) [52, 53, 54, 55]. Similarly, baseline counts of hematopoietic stem cells (HSCs), EPCs, and expression of CXCR4 and CXCR7 were significantly lower at rest in T1D group compared to healthy control group [56, 57]. Older adults demonstrated endothelium-dependent dilation of the brachial artery when compared to young healthy individuals. While there was no differences in the number of circulating EPCs; lower survival, migration, and proliferation was observed suggesting functional impairment of EPCs in older adults [58].

The level of circulating CD34+/KDR+ EPCs predicts the occurrence of cardiovascular events and death from cardiovascular causes and may help identify patients at increased cardiovascular risk [59]. Using 1751 individuals from the Framingham Offspring cohort there was an inverse association between CD34+ circulating progenitor cells (CPCs) and all-cause mortality when adjusting for standard risk factors. CD34+CD133+ CPCs were inversely associated with cardiovascular mortality [60]. Associations of CD34+ and CD34+CD133+ with mortality were strongest in participants with pre-existing CVD. Similarly, CD34+ and CD34+/KDR+ are observed to be significantly reduced in individuals with T2D while CD34+ cells only were also reduced in prediabetic individuals [61]. Post-challenge glucose was found to be an independent determinant of the levels of both CD34+ and CD34+/KDR+ in individuals with T2D and pre-diabetes [61]. Number of circulating EPCs and the combined Framingham risk factor score were found to be strongly correlated when assessed in healthy men [62]. In addition, flow-mediated brachial-artery reactivity was significantly related to endothelial function and number of progenitor cells [62]. Importantly, the level of circulating EPCs were a better predictor of vascular reactivity than was the presence or absence of conventional risk factors in healthy men [62]. EPC counts have been found to differ between stroke patients and control subjects, with EPC count being lower in stroke patients, independent of age [63]. The level of EPCs in stroke patients was also significantly correlated with the Framingham coronary score [63]. End stage kidney disease (ESKD) patients showed markedly decreased numbers of EPCs and colonies when compared with controls [64, 65]. ESKD had a decrease in EPC migratory function in response to VEGF and decrease in EPC incorporation into human umbilical vein endothelial cells. Framingham’s risk factor score of both ESKD and control group significantly correlated with the number of EPCs. Even in patients with mild stages of chronic kidney disease, EPC-mediated endogenous vascular regeneration has been shown to be impaired [66]. Thus, it appears both aging and chronic disease attenuate stem cell quantity and function, and are predictive of future adverse outcomes (Fig. 1).

Fig. 1.

Impact of aging, obesity, and disease on endothelial progenitor (EPCs) and mesenchymal stromal cell number (MSCs) and/or function and the potential for adverse cardiovascular events and all-cause mortality. Stimulus; aging, obesity; Source, bone marrow, umbilical cord blood, adipose tissue, pancreatic islet; Response, decrease in EPC and MSC number and/or function; Outcome, adverse cardiovascular events, all-cause mortality.

4. Current Physical Activity and Exercise Recommendations for Adults

Physical activity and exercise are essential for the maintenance and improvement of health and function [24, 25, 26]. Physical activity is defined as any bodily movement produced by skeletal muscles that results in an increase in caloric requirements above resting energy expenditure [67]. Exercise refers to a specific type of physical activity consisting of planned, structured, and repetitive bodily movement to improve and or maintain one or more components of physical fitness [67]. Aerobic activities, which consist of repetitive rhythmic movements such as walking, running, cycling, and swimming, are most often prescribed for targeting cardiovascular health. Muscle-strengthening activities (i.e., resistance exercise) include weight machines, free weights, resistance bands, and body-weight exercise and are prescribed for neuromuscular health. For older adults and adults living with chronic diseases, engaging in at least 150 minutes (2 hours and 30 minutes) to 300 minutes (5 hours) per week of moderate-intensity, or 75 minutes (1 hour and 15 minutes) to 150 minutes (2 hours and 30 minutes) a week of vigorous-intensity aerobic physical activity, or an equivalent combination of moderate- and vigorous-intensity aerobic physical activity is recommended [19, 67]. In addition, adults are encouraged to combine aerobic activity with 2 or more days per week of muscle-strengthening activities of moderate or greater intensity involving all major muscle groups [19, 67]. While meeting the physical activity and exercise recommendations are preferred, simply increasing the amount of physical activity and exercise above sedentary levels will confer health benefits even if the recommended guidelines are not achieved [68, 69]. Despite the importance of physical activity and exercise for maintaining or improving overall health, limited data exists on the effects of physical activity and exercise on EPCs and MSCs.

5. Acute Effects of Exercise on EPCs and MSCs

Studies investigating the responses of EPCs and MSCs show transient increases in cell numbers in response to exercise (Fig. 2) [29, 70, 71, 72]. Ferentinos et al. [70] conducted a systematic review and meta-analysis examining acute effects of different forms of exercise on circulating EPCs in healthy populations and found that prolonged endurance and resistance exercise had the most profound effect on circulating EPCs followed by maximal exercise. In another systematic review and meta-analysis examining healthy adults it was demonstrated that the extent and time-course of exercise-induced mobilization of circulating stem and progenitor cells differed between stem cell-subpopulations; i.e., EPCs, HSCs, and MSCs [71]. Early and non-specified stem cells were shown to increase significantly immediately after the initiation of exercise (i.e., 0–5 minutes) until 30-minutes post-exercise [71]. For EPC numbers, a significant increase was found until 12-48 hours after exercise and for HSC numbers at 0-5 minutes and at 3 hours after exercise [71]. No effect of exercise on MSC numbers was observed [71]. Importantly, these findings were not influenced by sex, intensity, or duration of the interventions assessed [71].

Fig. 2.

Effects of exercise on endothelial progenitor (EPCs) and mesenchymal stromal cells (MSCs) and the associated physiological adaptations. Stimulus; resistance exercise, cycling, running; Source, bone marrow, umbilical cord blood, adipose tissue, pancreatic islet; Response, increase in EPC and MSC number and/or function; Adaptation, angiogenesis, capillarization, skeletal muscle tissue repair.

In young healthy trained men, circulating EPC and serum concentrations of vascular endothelial growth factors (VEGF-A, VEGF-C, and VEGF-D), granulocyte colony stimulating factor, soluble Tie-2, soluble fms-like tyrosine kinase-1, and matrix metalloproteinases (MMP-1, MMP-2, MMP-3, MMP-9, and MMP-10) were higher in the postexercise period following a muscular endurance resistance exercise program (three circuits of 15-repetitions of six exercises) [73]. Circulating EPC were unchanged at 10-minutes postexercise but higher at 2-hours postexercise while the concentration of most angiogenic factors and metalloproteinases were higher at 10-minutes postexercise [73]. In young healthy women, resistance exercise using intensities of 60%, 70% and 80% 1-repetition maximum performed for 3 sets of 12-repetitions increased circulating EPC and levels of VEGF, hypoxia-inducible factor 1-alpha (HIF-1α), and erythropoietin after exercise with the change in EPCs from baseline being greatest in the 80% 1-RM group reaching the highest levels at 6 hours post-exercise [74]. The change in EPCs from baseline to 6 hours post-exercise was correlated with the change in VEGF and HIF-1α [74].

6. Chronic Effects of Exercise on EPCs and MSCs

Moderate levels of physical activity (50–70% of maximal heart rate for 20-minutes daily for 5 days per week) increase circulating numbers of EPCs and increases EPC colony count formation which appears to have an anti-apoptotic effect in subjects with pre-diabetes [75]. Such an effect of EPC is likely to depend on the degree of inflammation, hyperglycemia, and intrinsic antioxidant enzyme presence in the EPC. Similarly, it is also reported that exercise helps to reduce apoptosis that is mediated by phosphatidylinositol 3 (PI3)-kinase pathway which is dependent on nitric oxide bioavailability [76]. Prostaglandin E1 (PGE1)-mediated upregulation of EPC is also linked to the improvement of EPC function and improved angiogenesis [77]. The improvement of EPC number may be related to, and even preceded by, an increase in plasma VEGF [44]. For example, in patients with CAD, exercise induces a short-term cellular ischemia which increases HIF which in turn increases EPC number (dependent on VEGF and HIF) [44].

Exercise-induced osteogenesis are observed for bone marrow-derived MSCs [78]. Data from our laboratoryrevealed that exercise promotes osteogenic differentiation of fat derived MSCs in prediabeteic Veteran’s [79]. Interestingly, bone differentiation markers including RUNX genes, alkaline phosphatase (ALPL), and osteocalcin were significantly upregulated indicating osteogenic differentiation [79]. Cook et al. [80] discussed how signaling pathways manipulate MSC differentiation. Both bone morphogenic protein (BMP) and WNT signaling pathways play an important role in MSC differentiation towards bone formation following exercise. WNT signaling promotes osteogenic differentiation by upregulating RUNX (an important gene associated with bone formation) and by inhibiting peroxisome proliferation-activated receptor gamma (PPARᵞ) (a gene that promotes adipogenesis). On the other hand, BMPs activate osteogenic differentiation by activating a prominent bone forming transcription factor, RUNX2 [80]. Maredziak et al. [81] also showed that four-week old male C57B1/6 mice, 5-weeks of treadmill exercise increases bone marrow derived MSC number. Markers associated with osteogenesis (i.e., ALPL activity, osteopontin, and osteocalcin) were also found to be increased postexercise [81].

Collectively, exercise facilitates EPC differentiation and production from bone marrow and also helps to promote migration and homing-in of the progenitor cells to the hypoxic and damaged tissue which in turn improve angiogenesis and vasculogenesis [82]. As might be expected, the observed responses of increased count following exercise wane over time. Duration of exercise undertaken on a daily basis influences circulating EPCs numbers [44]. It has been reported that intensive and moderate exercise activity for 30-minutes increases circulating EPC number. However, this outcome is not seen when the time of exercise is reduced to 10 minutes [45]. It has also been demonstrated that a maximal bout of exercise stimulates a significant shift in CD34+ cells toward CD34+/KDR+ cells, indicating a shift of undifferentiated CD34+ progenitor cell moving towards a differentiated cell with endothelium-like characteristics.

7. Implications of Exercise on EPCs and MSCs in Aging and Chronic Disease

In older adults and clinical populations, physical activity and exercise elicit beneficials effects on endothelial function which may be explained, in part, by responses of EPCs and MSCs [83, 84]. Physical activity was associated with CD34+ CPCs only in individuals with CVD, a relationship that was maintained after adjustment for confounding variables [62]. Acute exercise promotes increases in stem cell numbers in older adults, however, the magnitude of response appears to be attenuated [53, 54]. In chronic heart failure patients (CHF), EPC mobilization was acutely increased after high intensity interval training or moderate intensity continuous exercise training, while findings were inconclusive after maximal exercise testing performed on a cycle ergometer [85]. In CHF patients, CD34+/KDR+ EPC numbers increased within 10-minutes following graded-exercise testing and remained elevated for up to 2 hours post-exercise [86]. The initial increase was small in the CHF patients and normalized within 30 minutes. However, the evolution of CD34+/KDR+ EPC numbers over time following graded-exercise testing overall was attenuated in CHF when compared to healthy controls. Exercise influenced SDF-1alpha levels over time without relation to changes in CD34+/KDR+ EPC. Maximal exercise tests acutely increased EPCs in ischemic or revascularized CAD [85]. In PAD patients, EPC levels increased up to 24 hours post-exercise while EPC mobilization was blunted after a single exercise session in patients with compromised metabolic health [85].

Intravascular ultrasound-based studies have shown atherosclerotic plaque reduction or retraction in response to exercise [87]. It has been reported that supervised exercise sessions boost the circulating EPC counts while increasing angiogenesis and improved endothelial function thereby decreasing the incidence of atherosclerosis [88, 89, 90]. In patients with hypertension, exercise reduces dysfunction of EPC, which promotes neovascularization and improves hypertension [91]. Mechanical stress to the tissue and vasculature is posited as a primary mechanism underlying the promotion of enhanced EPC function following exercise [82]. The mechanical force resulting from exercise directly or indirectly helps in improvement of EPC number and function [82]. Chronic moderate-intensity continuous exercise is shown to have a positive effect on circulating EPCs in older sedentary individuals which was accompanied by improvements in endothelial function and arterial stiffness [70]. In response to 8-weeks of cycling exercise at 65–85% heart rate reserve (HRR), there was no effect on baseline or exercise-induced numbers of HSCs and EPCs [54, 92]. Habitual physical activity in patients with CAD is associated with higher flow-mediated dilation and EPC count [93]. However, flow-mediated dilation was only related to increased habitual physical activity levels but not EPC count [93]. Patients with type I diabetes (T1D) performed 45-minutes of incline walking at 60% maximal oxygen consumption (VO2max). In both groups exercise increased circulating angiogenic cells however the increases were largely attenuated in the T1D group [56]. In young men with T1D , exercise did not induce changes in EPCs whereas in controls EPCs decreased after aerobic exercise and increased after resistance exercise [57]. Blood flow increased and vascular resistance decreased after resistance exercise in both groups. Reactive hyperemia increased 10-minutes after aerobic and resistance exercise in patients with T1D and controls with no differences between groups [57]. Despite increased vascular reactivity in both groups, EPCs were only affected by exercise in the controls which may indicate a blunted endothelium regenerating capacity in those with T1D [57]. Our laboratory has shown that 6-weeks of aerobic exercise improves EPC number and function, and upregulates endothelial cell-based gene expressions of critical endothelial specific genes such as e-NOS and VEGF while reducing inflammatory markers in patients with pre-diabetes [75].

Studies have showed that, similar to observations of EPCs, exercise may facilitate MSC homing into the site of injury [94]. It was reported that exercise induce homing of MSCs to extramedullary sites [95]. Often, the effect of MSC integration with host tissue and subsequent repair, increase with exercise [96]. It has also been reported that exercise increased the efficiency of MSC transplantation in cerebral ischemia in rats by inhibiting apoptosis [97]. Another study showed stromal vascular fraction, a well-known mixed population enriched with MSCs, when combined with exercise, together, help to improve pain in patients with knee osteoarthritis (OA) thereby establishing the synergistic effect of cell therapy and exercise in healing a common joint problem such as OA [98, 99]. Exercise also plays a vital role in differentiation of multipotent MSCs towards various adult tissues. MSCs can differentiate into bone, muscle, cartilage and adipose tissue depending on the need of the body’s reparative process. The differentiation is also dependent on the cellular environment.

8. Future Directions

The use of ASCs as a means of assessing and monitor health status is an evolving area and shows promise for the advancement of clinical care. However, several questions remain before its application can truly be appreciated. The lack of standardized designations of cells creates challenges for comparing findings across different research laboratories. The definition of EPC need to carefully delineated based on cell surface markers rather than cell type using phenotypic nomenclatures. Following appropriate cell surface-based designations, functional assays and there related reviews, can then be focused on. While increases in circulating ASCs are seen in response to both aerobic and resistance exercise, the exact dosing of such interventions has yet to be determined. Moreover, the time course of local and systemic adaptations associated with ASC response to exercise in aging and clinical populations requires further investigation. Additionally, given that most patients are prescribe one or more medications depending on their existing conditions, it will be essential to determine how ASCs respond to exercise interventions in the presence of specific medications.

9. Conclusions

ASCs, such as EPCs and MSCs, can act as a cellular biomarker for cardiovascular diseases, metabolic diseases, chronic rheumatological diseases, and infectious diseases. Use of ASCs as a biomarker have the potential to revolutionize the management of patients with a variety of metabolic and obesity related disorders and also pro-inflammatory diseases. Exercise offers beneficial effects on the proliferation and migratory function of EPCs and MSCs. Further investigation of clinical entities are urgently needed to understand the implications of interventions such as exercise, diet, and various medications on EPC and MSC quantity and function in aging and clinical populations.

Abbreviations

NIDDK, National Institute of Diabetes and Digestive and Kidney Diseases; WHO, World Health Organization; CDC; Centers for Disease Control and Prevention; CAD, coronary artery disease; T2D, type 2 diabetes; CVD, cardiovascular disease; METs, metabolic equivalence; hs-CRP, high-sensativity C-reactive protein; FDA, U.S. Food and Drug Administration; ASC, adult stem cells; HSC, hematopoietic stem cells; EPC, endothelial progenitor cell; MSC, mesenchymal stromal cell; KDR+, CD34+ plus kinase domain receptor; VEGFR-2, vascular endothelial growth factor-receptor-2; e-NOS, endothelial nitric oxide synthase; vWF, von-Willebrand’s factor; CPC, circulating progenitor cell; ESKD, end-stage kidney disease; MMP, matrix metalloproteinases; HIF-1α, hypoxia-inducible factor 1-alpha; ALPL, alkaline phosphatase; PPARᵞ, peroxisome proliferation-activated receptor gamma; BMP, bone morphogenic protein; CXCR4-expressing CACs, chemokine receptor 4 expressing circulating angiogenic cells; CHF, chronic heart failure; HRR, heart rate reserve; VO2max, maximal oxygen consumption; T1D, type 1 diabetes; OA, osteoarthritis.

Author Contributions

SS proposed the conceptualization of the research study. SS and JMG performed the literature review. SS and JMG synthesized the existing data. SS and JMG wrote the manuscript draft. SS and JMG contributed to editorial changes in the manuscript. SS and JMG read and approved the final manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This work was partially supported by the Department of Veterans Affairs Office of Research & Development, Rehabilitation Research & Development Career Development Award (CDA-2; 1IK2RX003423-01) (JMG).

Conflict of Interest

The authors declare no conflict of interest.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References
[1]
World Health Organization (WHO). Factsheet: Diabetes. 2021. Available at: http://www.who.int/mediacentre/factsheets/fs312/en/ (Accessed: 11 September 2022).
[2]
Center for Disease Control and Prevention (CDC). National Diabetes Statistics Report website. 2017. Available at: https://www.cdc.gov/diabetes/data/statistics/statistics-report.html (Accessed: 12 September 2022).
[3]
Center for Disease Control and Prevention (CDC). High Blood Pressure: Facts About Hypertension. 2016. vailable at: https://www.cdc.gov/bloodpressure/facts.htm (Accessed: 12 September 2022).
[4]
Center for Disease Control and Prevention (CDC). Stroke Facts. 2017. Available at: https://www.cdc.gov/stroke/facts.htm (Accessed: 12 September 2022).
[5]
Center for Disease Control and Prevention (CDC). Heart Disease Facts. 2017. Available at: https://www.cdc.gov/heartdisease/facts.htm (Accessed: 12 September 2022).
[6]
American Diabetes Association. Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care. 2018; 41: 917–928.
[7]
Booth FW, Roberts CK, Thyfault JP, Ruegsegger GN, Toedebusch RG. Role of Inactivity in Chronic Diseases: Evolutionary Insight and Pathophysiological Mechanisms. Physiological Reviews. 2017; 97: 1351–1402.
[8]
Booth FW, Roberts CK, Laye MJ. Lack of Exercise Is a Major Cause of Chronic Diseases. In Terjung R, (ed.) Comprehensive Physiology. John Wiley & Sons, Inc.: Hoboken, NJ, USA. 2012; 2: 1143–1211.
[9]
Kraus WE, Bittner V, Appel L, Blair SN, Church T, Després J-P, et al. The National Physical Activity Plan: A Call to Action From the American Heart Association: A Science Advisory From the American Heart Association. Circulation. 2015; 131: 1932–1940.
[10]
Lavie CJ, Ozemek C, Carbone S, Katzmarzyk PT, Blair SN. Sedentary Behavior, Exercise, and Cardiovascular Health. Circulation Research. 2019; 124: 799–815.
[11]
Myers J, McAuley P, Lavie CJ, Despres J-P, Arena R, Kokkinos P. Physical Activity and Cardiorespiratory Fitness as Major Markers of Cardiovascular Risk: Their Independent and Interwoven Importance to Health Status. Progress in Cardiovascular Diseases. 2015; 57: 306–314.
[12]
Katzmarzyk PT, Powell KE, Jakicic JM, Troiano RP, Piercy K, Tennant B. Sedentary Behavior and Health: Update from the 2018 Physical Activity Guidelines Advisory Committee. Medicine & Science in Sports & Exercise. 2019; 51: 1227–1241.
[13]
Myers J, Vainshelboim B, Kamil-Rosenberg S, Chan K, Kokkinos P. Physical Activity, Cardiorespiratory Fitness, and Population-Attributable Risk. Mayo Clinic Proceedings. 2021; 96: 342–349.
[14]
Martinez-Gomez D, Cabanas-Sanchez V, Yu T, Rodriguez-Artalejo F, Ding D, Lee I, et al. Long-term leisure-time physical activity and risk of all-cause and cardiovascular mortality: dose–response associations in a prospective cohort study of 210 327 Taiwanese adults. British Journal of Sports Medicine. 2022; 56: 919–926.
[15]
Kokkinos P, Faselis C, Samuel IBH, Pittaras A, Doumas M, Murphy R, et al. Cardiorespiratory Fitness and Mortality Risk Across the Spectra of Age, Race, and Sex. Journal of the American College of Cardiology. 2022; 80: 598–609.
[16]
Kerr NR, Booth FW. Contributions of physical inactivity and sedentary behavior to metabolic and endocrine diseases. Trends in Endocrinology & Metabolism. 2022; 33: 817–827.
[17]
Myers J, Kaykha A, George S, Abella J, Zaheer N, Lear S, et al. Fitness versus physical activity patterns in predicting mortality in men. The American Journal of Medicine. 2004; 117: 912–918.
[18]
Artero EG, Lee D, Lavie CJ, España-Romero V, Sui X, Church TS, et al. Effects of muscular strength on cardiovascular risk factors and prognosis. Journal of Cardiopulmonary Rehabilitation and Prevention. 2012; 32: 351–358.
[19]
U.S. Department of Health and Human Services. Physical Activity Guidelines for Americans. 2nd edn. Washington, DC. 2018.
[20]
Bennie JA, De Cocker K, Teychenne MJ, Brown WJ, Biddle SJH. The epidemiology of aerobic physical activity and muscle-strengthening activity guideline adherence among 383,928 U.S. adults. International Journal of Behavioral Nutrition and Physical Activity. 2019; 16: 34.
[21]
Kraus WE, Powell KE, Haskell WL, Janz KF, Campbell WW, Jakicic JM, et al. Physical Activity, all-Cause and Cardiovascular Mortality, and Cardiovascular Disease. Medicine & Science in Sports & Exercise. 2019; 51: 1270–1281.
[22]
Schwarz PE, Greaves CJ, Lindström J, Yates T, Davies MJ. Nonpharmacological interventions for the prevention of type 2 diabetes mellitus. Nature Reviews Endocrinology. 2012; 8: 363–373.
[23]
Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Reduction in the Incidence of Type 2 Diabetes with Lifestyle Intervention or Metformin. New England Journal of Medicine. 2002; 346: 393–403.
[24]
Center for Disease Control and Prevention (CDC). Benefits of Physical Activity. 2015. Available at: https://www.cdc.gov/physicalactivity/basics/pa-health/index.htm (Accessed: 12 September 2022).
[25]
González-Gross M. Biomarkers of Physical Activity and Exercise. Nutricion Hospitalaria. 2015; 31 Suppl 3: 237–244.
[26]
Zhao M, Veeranki SP, Magnussen CG, Xi B. Recommended physical activity and all cause and cause specific mortality in US adults: prospective cohort study. British Medical Journal. 2020; 370 : m2031.
[27]
Naci H, Ioannidis JPA. Comparative effectiveness of exercise and drug interventions on mortality outcomes: metaepidemiological study. British Medical Journal. 2013; 347: f5577.
[28]
De Biase C, De Rosa R, Luciano R, De Luca S, Capuano E, Trimarco B, et al. Effects of physical activity on endothelial progenitor cells (EPCs). Frontiers in Physiology. 2014; 4: 414.
[29]
Boppart MD, De Lisio M, Witkowski S. Exercise and Stem Cells. Progress in Molecular Biology and Translational Science. 2015; 7: 423–456.
[30]
Witkowski S, Jenkins NT, Hagberg JM. Enhancing Treatment for Cardiovascular Disease: exercise and circulating angiogenic cells. Exercise and Sport Sciences Reviews. 2011; 39: 93–101.
[31]
Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nature Reviews Immunology. 2008; 8: 726–736.
[32]
Minguell JJ, Erices A, Conget P. Mesenchymal Stem Cells. Experimental Biology and Medicine. 2001; 226: 507–520.
[33]
Caplan AI. Mesenchymal stem cells. Journal of Orthopaedic Research. 1991; 9: 641–650.
[34]
Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. Journal of Cellular Biochemistry. 2006; 98: 1076–1084.
[35]
Roufosse CA, Direkze NC, Otto WR, Wright NA. Circulating mesenchymal stem cells. The International Journal of Biochemistry & Cell Biology. 2004; 36: 585–597.
[36]
Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, Krause DS, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006; 8: 315–317.
[37]
Domingues CC, Kundu N, Dore FJ, Sen S. Genetic Modification of Stem Cells in Diabetes and Obesity. In Jamal F, (ed.) Genetic Engineering - An Insight into the Strategies and Applications. IntechOpen: London, United Kingdom. 2016.
[38]
Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Engineering. 2001; 7: 211–228.
[39]
Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, Hérault O, et al. The in vitro migration capacity of human bone marrow mesenchymal stem cells: comparison of chemokine and growth factor chemotactic activities. Stem Cells. 2007; 25: 1737–1745.
[40]
Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Communication and Signaling. 2011; 9: 12.
[41]
Galderisi U, Peluso G, Di Bernardo G. Clinical Trials Based on Mesenchymal Stromal Cells are Exponentially Increasing: where are we in Recent Years? Stem Cell Reviews and Reports. 2022; 18: 23–36.
[42]
Boppart MD, De Lisio M, Zou K, Huntsman HD. Defining a role for non-satellite stem cells in the regulation of muscle repair following exercise. Frontiers in Physiology. 2013; 4: 310.
[43]
Bourzac C, Bensidhoum M, Pallu S, Portier H. Use of adult mesenchymal stromal cells in tissue repair: impact of physical exercise. American Journal of Physiology-Cell Physiology. 2019; 31: C642–C654.
[44]
Adams V, Lenk K, Linke A, Lenz D, Erbs S, Sandri M, et al. Increase of circulating endothelial progenitor cells in patients with coronary artery disease after exercise-induced ischemia. Arteriosclerosis, Thrombosis, and Vascular Biology. 2004; 24: 684–690.
[45]
Laufs U, Urhausen A, Werner N, Scharhag J, Heitz A, Kissner G, et al. Running exercise of different duration and intensity: effect on endothelial progenitor cells in healthy subjects. European Journal of Cardiovascular Prevention & Rehabilitation. 2005; 12: 407–414.
[46]
Awal HB, Nandula SR, Domingues CC, Dore FJ, Kundu N, Brichacek B, et al. Linagliptin, when compared to placebo, improves CD34+ve endothelial progenitor cells in type 2 diabetes subjects with chronic kidney disease taking metformin and/or insulin: a randomized controlled trial. Cardiovascular Diabetology. 2020; 19: 72.
[47]
Nandula SR, Kundu N, Awal HB, Brichacek B, Fakhri M, Aimalla N, et al. Role of Canagliflozin on function of CD34+ve endothelial progenitor cells (EPC) in patients with type 2 diabetes. Cardiovascular Diabetology. 2021; 20: 44.
[48]
Elzarki AF, Nandula SR, Awal H, Simon GL, Sen S. Cardiovascular disease (CVD) risk assessment of HIV medication regimens using hematopoietic CD34+ progenitor cells. Stem Cell Research & Therapy. 2022; 13: 103.
[49]
Moreno PR, Sanz J, Fuster V. Promoting Mechanisms of Vascular Health. Journal of the American College of Cardiology. 2009; 53: 2315–2323.
[50]
Bakogiannis C, Tousoulis D, Androulakis E, Briasoulis A, Papageorgiou N, Vogiatzi G, et al. Circulating Endothelial Progenitor Cells as Biomarkers for Prediction of Cardiovascular Outcomes. CMC. 2012; 19: 2597–2604.
[51]
Williamson K, Stringer SE, Alexander MY. Endothelial Progenitor Cells Enter the Aging Arena. Frontiers in Physiology. 2012; 3: 30.
[52]
Shaffer RG, Greene S, Arshi A, Supple G, Bantly A, Moore JS, et al. Effect of acute exercise on endothelial progenitor cells in patients with peripheral arterial disease. Vascular Medicine. 2006; 11: 219–226.
[53]
Ross MD, Malone EM, Simpson R, Cranston I, Ingram L, Wright GP, et al. Lower resting and exercise-induced circulating angiogenic progenitors and angiogenic T cells in older men. American Journal of Physiology-Heart and Circulatory Physiology. 2018; 314: H392–H402.
[54]
Thijssen DHJ, Vos JB, Verseyden C, van Zonneveld AJ, Smits P, Sweep FCGJ, et al. Haematopoietic stem cells and endothelial progenitor cells in healthy men: effect of aging and training. Aging Cell. 2006; 5: 495–503.
[55]
Ross MD, Malone E, Florida-James G. Vascular Ageing and Exercise: Focus on Cellular Reparative Processes. Oxidative Medicine and Cellular Longevity. 2016; 2016: 1–15.
[56]
Taylor GS, Shaw A, Smith K, Capper TE, Scragg JH, Cronin M, et al. Type 1 diabetes patients increase CXCR4+ and CXCR7+ haematopoietic and endothelial progenitor cells with exercise, but the response is attenuated. Scientific Reports. 2021; 11: 14502.
[57]
Waclawovsky G, Umpierre D, Figueira FR, De Lima ES, Alegretti AP, Schneider L, et al. Exercise on Progenitor Cells in Healthy Subjects and Patients with Type 1 Diabetes. Medicine & Science in Sports & Exercise. 2016; 48: 190–199.
[58]
Heiss C, Keymel S, Niesler U, Ziemann J, Kelm M, Kalka C. Impaired Progenitor Cell Activity in Age-Related Endothelial Dysfunction. Journal of the American College of Cardiology. 2005; 45: 1441–1448.
[59]
Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, et al. Circulating Endothelial Progenitor Cells and Cardiovascular Outcomes. New England Journal of Medicine. 2005; 353: 999–1007.
[60]
Muggeridge D, Dodd J, Ross MD. CD34+ progenitors are predictive of mortality and are associated with physical activity in cardiovascular disease patients. Atherosclerosis. 2021; 333: 108–115.
[61]
Fadini GP, Pucci L, Vanacore R, Baesso I, Penno G, Balbarini A, et al. Glucose tolerance is negatively associated with circulating progenitor cell levels. Diabetologia. 2007; 50: 2156–2163.
[62]
Hill JM, Zalos G, Halcox JPJ, Schenke WH, Waclawiw MA, Quyyumi AA, et al. Circulating Endothelial Progenitor Cells, Vascular Function, and Cardiovascular Risk. New England Journal of Medicine. 2003; 348: 593–600.
[63]
Ghani U, Shuaib A, Salam A, Nasir A, Shuaib U, Jeerakathil T, et al. Endothelial Progenitor Cells during Cerebrovascular Disease. Stroke. 2005; 36: 151–153.
[64]
Choi J, Kim KL, Huh W, Kim B, Byun J, Suh W, et al. Decreased Number and Impaired Angiogenic Function of Endothelial Progenitor Cells in Patients with Chronic Renal Failure. Arteriosclerosis, Thrombosis, and Vascular Biology. 2004; 24: 1246–1252.
[65]
Schlieper G, Hristov M, Brandenburg V, Krüger T, Westenfeld R, Mahnken AH, et al. Predictors of low circulating endothelial progenitor cell numbers in haemodialysis patients. Nephrology Dialysis Transplantation. 2008; 23: 2611–2618.
[66]
Jie KE, Zaikova MA, Bergevoet MWT, Westerweel PE, Rastmanesh M, Blankestijn PJ, et al. Progenitor cells and vascular function are impaired in patients with chronic kidney disease. Nephrology Dialysis Transplantation. 2010; 25: 1875–1882.
[67]
American College of Sports Medicine. In Liguori G, Feito Y, Fountaine C, Roy B, (eds.) ACSM’s guidelines for exercise testing and prescription. 11th edn. Wolters Kluwer: Philadelphia. 2021.
[68]
Myers J, Kokkinos P, Arena R, LaMonte MJ. The impact of moving more, physical activity, and cardiorespiratory fitness: why we should strive to measure and improve fitness. Progress in Cardiovascular Diseases. 2021; 64: 77–82.
[69]
Franklin BA, Eijsvogels TMH, Pandey A, Quindry J, Toth PP. Physical activity, cardiorespiratory fitness, and cardiovascular health: A clinical practice statement of the ASPC Part I: Bioenergetics, contemporary physical activity recommendations, benefits, risks, extreme exercise regimens, potential maladaptations. American Journal of Preventive Cardiology. 2022; 12: 100424.
[70]
Ferentinos P, Tsakirides C, Swainson M, Davison A, Martyn-St James M, Ispoglou T. The impact of different forms of exercise on endothelial progenitor cells in healthy populations. European Journal of Applied Physiology. 2022; 122: 1589–1625.
[71]
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 Reviews and Reports. 2021; 17: 1091–1120.
[72]
Möbius-Winkler S, Hilberg T, Menzel K, Golla E, Burman A, Schuler G, et al. Time-dependent mobilization of circulating progenitor cells during strenuous exercise in healthy individuals. Journal of Applied Physiology. 2009; 107: 1943–1950.
[73]
Ross MD, Wekesa AL, Phelan JP, Harrison M. Resistance Exercise Increases Endothelial Progenitor Cells and Angiogenic Factors. Medicine & Science in Sports & Exercise. 2014; 46: 16–23.
[74]
Ribeiro F, Ribeiro IP, Gonçalves AC, Alves AJ, Melo E, Fernandes R, et al. Effects of resistance exercise on endothelial progenitor cell mobilization in women. Scientific Reports. 2017; 7: 17880.
[75]
Sen S, Witkowski S, Lagoy A, Islam AM. A Six-Week Home Exercise Program Improves Endothelial Function and CD34 + Circulating Progenitor Cells in Patients With Pre-Diabetes. Journal of Endocrinology and Metabolism. 2015; 5: 163–171.
[76]
Suhr F, Brenig J, Müller R, Behrens H, Bloch W, Grau M. Moderate Exercise Promotes Human RBC-NOS Activity, NO Production and Deformability through Akt Kinase Pathway. PLoS ONE. 2012; 7: e45982.
[77]
Gensch C, Clever Y, Werner C, Hanhoun M, Böhm M, Laufs U. Regulation of endothelial progenitor cells by prostaglandin E1 via inhibition of apoptosis. Journal of Molecular and Cellular Cardiology. 2007; 42: 670–677.
[78]
Liu S-Y, He Y-B, Deng S-Y, Zhu W-T, Xu S-Y, Ni G-X. Exercise affects biological characteristics of mesenchymal stromal cells derived from bone marrow and adipose tissue. International Orthopaedics (SICOT). 2017; 41: 1199–209.
[79]
Kundu N, Domingues CC, Nylen ES, Paal E, Kokkinos P, Sen S. Endothelium-Derived Factors Influence Differentiation of Fat-Derived Stromal Cells Post-Exercise in Subjects with Prediabetes. Metabolic Syndrome and Related Disorders. 2019; 17: 314–322.
[80]
Cook D, Genever P. Regulation of Mesenchymal Stem Cell Differentiation. Transcriptional and Translational Regulation of Stem Cells. 2013; 16: 213–229.
[81]
Marędziak M, Śmieszek A, Chrząstek K, Basinska K, Marycz K. Physical Activity Increases the Total Number of Bone-Marrow-Derived Mesenchymal Stem Cells, Enhances Their Osteogenic Potential, and Inhibits Their Adipogenic Properties. Stem Cells International. 2015; 2015: 1–11.
[82]
Wahl P, Brixius K, Bloch W. Exercise‐induced stem cell activation and its implication for cardiovascular and skeletal muscle regeneration. Minimally Invasive Therapy & Allied Technologies. 2008; 17: 91–99.
[83]
Moyna NM, Thompson PD. The effect of physical activity on endothelial function in man. Acta Physiologica Scandinavica. 2004; 180: 113–123.
[84]
Jasperse JL, Laughlin MH. Endothelial Function and Exercise Training: Evidence from Studies Using Animal Models. Medicine & Science in Sports & Exercise. 2006; 38: 445–454.
[85]
Ferentinos P, Tsakirides C, Swainson M, Davison A, Martyn-St James M, Ispoglou T. The impact of different forms of exercise on circulating endothelial progenitor cells in cardiovascular and metabolic disease. European Journal of Applied Physiology. 2022; 122: 815–860.
[86]
Van Craenenbroeck EM, Bruyndonckx L, Van Berckelaer C, Hoymans VY, Vrints CJ, Conraads VM. The effect of acute exercise on endothelial progenitor cells is attenuated in chronic heart failure. European Journal of Applied Physiology. 2011; 111: 2375–2379.
[87]
Ajijola OA, Dong C, Herderick EE, Ma Q, Goldschmidt-Clermont PJ, Yan Z. Voluntary Running Suppresses Proinflammatory Cytokines and Bone Marrow Endothelial Progenitor Cell Levels in Apolipoprotein-E–Deficient Mice. Antioxidants & Redox Signaling. 2009; 11: 15–23.
[88]
Schlager O, Giurgea A, Schuhfried O, Seidinger D, Hammer A, Gröger M, et al. Exercise training increases endothelial progenitor cells and decreases asymmetric dimethylarginine in peripheral arterial disease: a randomized controlled trial. Atherosclerosis. 2011; 217: 240–248.
[89]
Van Craenenbroeck EM, Beckers PJ, Possemiers NM, Wuyts K, Frederix G, Hoymans VY, et al. Exercise acutely reverses dysfunction of circulating angiogenic cells in chronic heart failure. European Heart Journal. 2010; 31: 1924–1934.
[90]
Rehman J, Li J, Parvathaneni L, Karlsson G, Panchal VR, Temm CJ, et al. Exercise acutely increases circulating endothelial progenitor cells and monocyte-/macrophage-derived angiogenic cells. Journal of the American College of Cardiology. 2004; 43: 2314–2318.
[91]
Fernandes T, Nakamuta JS, Magalhães FC, Roque FR, Lavini-Ramos C, Schettert IT, et al. Exercise training restores the endothelial progenitor cells number and function in hypertension: implications for angiogenesis. Journal of Hypertension. 2012; 30: 2133–2143.
[92]
Lockard MM, Witkowski S, Jenkins NT, Spangenburg EE, Obisesan TO, Hagberg JM. Thrombin and exercise similarly influence expression of cell cycle genes in cultured putative endothelial progenitor cells. Journal of Applied Physiology. 2010; 108: 1682–1690.
[93]
Luk T, Dai Y, Siu C, Yiu K, Chan H, Fong DY, et al. Habitual physical activity is associated with endothelial function and endothelial progenitor cells in patients with stable coronary artery disease. European Journal of Cardiovascular Prevention & Rehabilitation. 2009; 16: 464–471.
[94]
Schmidt A, Bierwirth S, Weber S, Platen P, Schinkothe T, Bloch W. Short intensive exercise increases the migratory activity of mesenchymal stem cells. British Journal of Sports Medicine. 2009; 43: 195–198.
[95]
Emmons R, Niemiro GM, Owolabi O, De Lisio M. Acute exercise mobilizes hematopoietic stem and progenitor cells and alters the mesenchymal stromal cell secretome. Journal of Applied Physiology. 2016; 120: 624–632.
[96]
Shin MS, Park HK, Kim TW, Ji ES, Lee JM, Choi HS, et al. Neuroprotective Effects of Bone Marrow Stromal Cell Transplantation in Combination with Treadmill Exercise Following Traumatic Brain Injury. International Neurourology Journal. 2016; 20: S49–S56.
[97]
Zhang Y-X, Yuan M-Z, Cheng L, Lin L-Z, Du H-W, Chen R-H, et al. Treadmill exercise enhances therapeutic potency of transplanted bone mesenchymal stem cells in cerebral ischemic rats via anti-apoptotic effects. BMC Neuroscience. 2015; 16: 56.
[98]
Gibbs N, Diamond R, Sekyere EO, Thomas WD. Management of knee osteoarthritis by combined stromal vascular fraction cell therapy, platelet-rich plasma, and musculoskeletal exercises: a case series. Journal of Pain Research. 2015; 8: 799–806.
[99]
Aoyama T, Fujita Y, Madoba K, Nankaku M, Yamada M, Tomita M, et al. Rehabilitation program after mesenchymal stromal cell transplantation augmented by vascularized bone grafts for idiopathic osteonecrosis of the femoral head: a preliminary study. Archives of Physical Medicine and Rehabilitation. 2015; 96: 532–539.
Share
Back to top