Next Article in Journal / Special Issue
Next Steps for Intradialytic Cycling Research
Previous Article in Journal / Special Issue
Targeted Non-Pharmacological Interventions for People Living with Frailty and Chronic Kidney Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Exercise Training in Delaying Kidney Function Decline in Non-Dialysis-Dependent Chronic Kidney Disease

1
School of Human and Behavioural Sciences, Bangor University, Bangor LL57 2DG, UK
2
Renal Department, Ysbyty Gwynedd Hospital, Bangor LL57 2PW, UK
*
Authors to whom correspondence should be addressed.
Kidney Dial. 2022, 2(2), 262-286; https://doi.org/10.3390/kidneydial2020026
Submission received: 11 March 2022 / Revised: 7 April 2022 / Accepted: 8 May 2022 / Published: 25 May 2022
(This article belongs to the Special Issue Lifestyle Interventions to Prevent Kidney Diseases)

Abstract

:
Chronic Kidney Disease (CKD) is a progressive condition characterised by declining eGFR and associated, particularly in advanced stages, with increased morbidity and cardiovascular mortality. Current treatment options for delaying disease progression are limited to a small number of pharmacological agents. Considering that rates of kidney function decline are greater in patients with lower levels of habitual physical activity, there is interest in the potential benefits of structured exercise training in delaying CKD progression. This discursive review summarises the current state-of-play in this field of research by critically analysing the published systematic reviews of randomised controlled trials of structured exercise training in the non-dialysis CKD population. Several issues are highlighted that hamper definite conclusions as to the therapeutic effectiveness of exercise training for this purpose. However, following an overview of the pathophysiology and risk factors for kidney function decline, several potential mechanisms explaining how exercise training may benefit CKD progression are offered. Finally, suggestions for future research in this area are made. The review concludes that there is a need for further research on the effectiveness of exercise before it can be recommended as part of routine care for the purpose of delaying CKD progression. Exercise can be recommended, however, to individual patients because of a potential benefit to kidney function, and definite benefits to other outcomes such as quality of life, with no apparent evidence of harm.

1. Introduction

Chronic Kidney Disease (CKD) is a progressive condition characterised by variable, but usually inevitable, annual rates of estimated glomerular filtration rate (eGFR) decline [1]. Even before progressing to end-stage kidney disease (ESKD), CKD is associated with increased mortality, particularly cardiovascular death [2]. Furthermore, CKD is an independent risk factor for cardiovascular disease with an increased risk of death, cardiovascular events and hospitalisation as eGFR falls [3]. Current treatment options for delaying the progression of CKD are limited to a small handful of therapeutic options, namely renin-angiotensin-aldosterone (RAS) system inhibitors, sodium bicarbonate supplementation and sodium-glucose transporter 2 (SGLT-2) inhibitors [4,5]. In the hunt for additional strategies, exercise training has been considered a possible therapy for delaying disease progression.
Despite evidence of exercise-induced acute kidney injury (AKI) in endurance athletes and increased proteinuria post-exercise, concerns that exercise may have a detrimental effect on kidney function have been allayed by a wealth of studies showing exercise training to be safe [6,7,8,9]. Indeed, structured exercise training has been shown to have multiple beneficial effects in the non-dialysis-dependent CKD (ND-CKD) population, including improved aerobic capacity [10,11,12,13], physical function [9,14,15], muscle strength [14,15,16] and health-related quality of life (HRQoL) [9,17]. Increased habitual physical activity is associated with better survival in both ESKD [18] and ND-CKD patients [19,20] (though there is a possible influence of selection bias and other factors not adjusted for in these observational studies).
With regards to disease progression, there is observational data that links low physical activity with faster CKD progression. People with CKD are generally less active than the general population [21], with physical activity levels reduced further in advanced disease stages [22,23,24,25]. More than that, lower physical activity levels appear to be predictive of kidney function decline [26,27,28].
Therefore, understanding whether structured exercise can delay CKD progression will be of benefit to patients and practitioners. This narrative review briefly outlines the pathophysiology of kidney disease progression and highlights several key risk factors for kidney disease progression. The current evidence for the effect of exercise training on eGFR is reviewed before considering plausible ways in which exercise may help delay CKD progression. Future considerations for scientific studies are also suggested.

2. The Pathophysiology of CKD Progression

Whilst a thorough description of the pathophysiology of CKD progression is beyond the scope of this review and is available elsewhere (e.g., [29,30,31,32]), an outline of the key processes and mediators will aid the reader in understanding the potential beneficial effects of exercise training.
It is widely acknowledged that, regardless of the original aetiology of kidney disease, there are common subsequent pathways that ultimately lead to kidney fibrosis and loss of function [32,33,34,35]. Most kidney diseases begin in the glomeruli [32,36], with damage leading to several downstream effects. Firstly, there is a propagation of injury within the glomeruli, including to the podocytes, a key cell in the filtration barrier [37]. Podocyte injury results in increased passage of protein into the tubules, which in turn, results in increased protein uptake and processing by tubular cells. This stimulates proximal tubular cells into assuming a pro-inflammatory phenotype with altered transcription factor activity, including increased NF- κ B with a resultant production of a number of pro-inflammatory and pro-fibrotic mediators, including MCP-1 and TGF- β 1 [36,38]. Damage to glomeruli also leads to hyperfiltration of intact glomeruli in an attempt to compensate for the loss in number of functioning glomeruli [33,39]. This adaptive mechanism has negative consequences, including damage to glomerular cells and increased proteinuria. Furthermore, it compromises tubular blood supply both by glomerulosclerosis, with a consequent collapse of capillaries, and by haemodynamic changes (increased arteriolar resistance leading to reduced peritubular capillary blood flow) [32,35]. Hyperfiltration also leads to increased tubular activity, including the metabolically costly process of sodium reabsorption. The increased oxygen demand may lead to oxidative stress but also to relative cortical hypoxia [32,35]. Tissue hypoxia also results from capillary rarefaction: inflammatory processes in the tubulo-interstitium that result in a loss of endothelial integrity and cell death. The importance of a healthy endothelium has been highlighted by a number of experimental models which remove the key endothelial cell product nitric oxide (NO), produced by the enzyme endothelial nitric oxide synthase (eNOS): these include eNOS knockout diabetic mice, which show more rapid eGFR decline than wildtype [40]. The reduced blood supply caused by capillary rarefaction is exacerbated by the build-up of the interstitial fibrotic matrix (due to the inflammatory processes mentioned above), which increases the distance between surviving capillaries and the tubules they are supplying, thus worsening parenchymal hypoxia [41,42]. These various pathways of tubular injury lead to inflammatory processes that perpetuate further injury in all nephron compartments resulting in interstitial inflammation and fibrosis and loss of kidney function [36,43].
Driving these pathological processes is an imbalance of various mediators. Those briefly outlined here are amongst the most well-recognised and are relevant for this discussion about the potential effects of exercise. The importance of preserving tubular and glomerular endothelium has been highlighted. Loss of vascular endothelial growth factor (VEGF), produced by both podocytes and tubular epithelial cells and important for maintaining endothelial integrity, has been implicated in furthering the progression of kidney damage [41,44], though it should also be noted that aberrant VEGF activity may be fibrogenic [45]. Angiotensin II (AngII), originally known primarily as a haemodynamic actor, has pleiotropic effects and is a key mediating molecule of many pathological pathways of kidney disease progression, including increasing proteinuria through direct effects on the filtration barrier, stimulating key inflammatory pathways and inducing oxidative stress [46]. Oxidative stress, an imbalance in pro-oxidant and anti-oxidant molecules, has been shown in experimental models to contribute to CKD progression both by direct cellular damage and by stimulating pro-inflammatory processes [47,48]. Prominent imbalances in inflammatory pathways that promote kidney damage include increased expression of pro-inflammatory transcription factors, particularly NF- κ B [31,49], and broadly reduced anti-inflammatory Nrf2 (nuclear factor erythroid 2–related factor 2) [31]; chemokines, particularly MCP-1 (monocyte chemoattractant protein 1) and RANTES (Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted) [29,30]; and cytokines, including IL-1 β , Il-6, TNF- α [50,51]. Finally, sympathetic nervous system (SNS) overactivity, which is common in patients with CKD [52], has been implicated as a potential contributor not only of CKD-related hypertension, but also of disease progression [53,54], possibly by damaging peritubular capillaries leading to ischaemia [55].

3. Risk Factors for CKD Progression

There are several well-established risk factors for kidney disease progression, regardless of the original aetiology. General population studies [56,57,58] and observational studies in patients with established CKD [59,60,61] have demonstrated a marked worsening of renal outcomes in patients with hypertension; the risk increases as blood pressure increases. Additionally, there is evidence of benefit, i.e., slower eGFR decline, with improved blood pressure control in interventional studies [62,63]. In addition to hypertension per se, recent studies also identify that raised short-term systolic blood pressure variability (which is prevalent in patients with CKD [64] and partly due to increased sympathetic nervous activity [65]) is predictive of disease progression (rapid eGFR decline [66] or ESKD incidence [67]) in patients with established CKD.
The usual processes of renal blood flow autoregulation protect the glomerulus from systemic hypertension. Renal autoregulation, however, can be overwhelmed by sustained or severe hypertension [68,69]. Even in situations where autoregulation is still effective, hypertension can result in progressive renal damage in cases where there is underlying renal injury of another cause [69]. In both these situations, barotrauma, i.e., damage caused by pressure, results from persistent or severe hypertension. Examples of resultant damage include upregulation of fibrotic repair mechanisms due to capillary and mesangial cell stretching, worsening proteinuria (with its downstream effects) [70] and shedding or effacement of podocytes [71]. In addition to direct pressure effects, there is evidence that hypertension upregulates or exacerbates other pathogenic pathways, including endothelial dysfunction [72] and oxidative stress [73].
Similarly, obesity or raised body mass index (BMI) are strongly associated with adverse renal outcomes in general population studies and CKD cohorts [74,75,76]. Weight loss interventions, including surgical, pharmacological and lifestyle interventions, are able to prevent renal function decline and even improve eGFR in patients with CKD [77,78,79].
There are two broad mechanisms whereby obesity contributes to CKD progression. Adipocytes (fat cells) are not just storage cells, but produce various hormones, cytokines and other pro-inflammatory molecules, including angiotensinogen (the precursor to Ang II), TNF- α and IL-6 [80]. Altered levels of these molecules occur in obesity and may be contributory to renal damage [80]. Secondly, both animal and human studies have demonstrated that BMI in overweight or obese categories leads to increased renal plasma flow and glomerular hyperfiltration [81]. As discussed above, hyperfiltration is linked to progressive glomerular damage and proteinuria.
Diabetes is the most common cause of CKD worldwide [82]; the presence of diabetes increases risk of ESKD and risk of incident moderate CKD in population studies [57,74,83], as well as increased risk of eGFR decline in observational studies in people with established CKD [84]. Glycaemic control has been established as a risk factor for diabetic kidney disease incidence and progression in randomised trials of strict control with various anti-glycaemic agents in both Type 1 [85] and Type 2 [86] diabetes mellitus.
The potential pathological effects of hyperglycaemia on the kidney are numerous and involve multiple cells of the nephron, particularly endothelial and mesangial cells. Hyperglycaemia stimulates various metabolic pathways, which are damaging when overwhelmed or inappropriately activated. These include stimulating TGF- β 1 production, both directly and by protein kinase-C pathway activation [87], and aberrant mitochondrial activity with resultant oxidative stress production [88]. Advanced glycation end-products (AGEs), the products of non-enzymatic reactions between glucose and proteins or lipids, also form readily in hyperglycaemic states and can independently stimulate similar pathological pathways [87]. Hyperglycaemia also impairs renal autoregulation, and so augments the harmful effects of hypertension in the glomerulus [89].
Even in non-diabetic patients, insulin resistance is common in CKD [90]. Insulin resistance is a consequence of visceral obesity [91] and is highly related to metabolic syndrome [92], but there are additional factors in CKD—namely inflammatory and oxidative stress pathways, as well as metabolic acidosis—which promotes insulin resistance [90]. Both metabolic syndrome and insulin resistance have been implicated as risk factors for CKD development and progression [93,94].
Insulin resistance leads to hyperinsulinaemia. Raised insulin concentration, acting via the insulin-like growth factor-1 (IGF-1) receptor, stimulates vascular smooth muscle cell proliferation, mesangial cell growth and reduced metalloproteinase activity; the result is an alteration of glomerular structure and renal fibrosis [90]. Insulin can also impair renal autoregulation by a sodium-retaining action in the renal tubules, potentially leading to increased glomerular pressure [90]. It is also likely that insulin resistance reduces NO availability, thus having detrimental effects on renal haemodynamics [95].
Understanding the importance of the pathophysiology and risk factors for CKD progression is important for establishing plausible therapeutic targets and interventions. As will be reviewed later, the risk factors discussed here are modifiable by exercise intervention. Next, we will review the evidence concerning exercise training on CKD progression itself.

4. The Current State of the Research on Exercise Training to Delay CKD Progression

There has been, for some time, interest in the potential beneficial effects of exercise training on CKD progression per se. The earliest randomised trial in humans found no benefit [96]. Some other early studies in this field, however, showed reduced rates of decline, and even improvement, after aquatic [97], aerobic [98] and resistance [99] exercise interventions.
Since then, numerous studies have been published, which include a measure of kidney function as an outcome of interest [10,11,12,13,15,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114]. The effect of exercise training on CKD progression (i.e., eGFR decline) has been further examined in several systematic reviews with meta-analysis [17,115,116,117,118,119], identified by a comprehensive literature search (see Supplementary Materials). These analyses are summarised in Table 1. As can be seen in Table 1, these reviews have reported conflicting findings, with almost equal numbers reporting a statistically significant, and clinically meaningful, beneficial effect on eGFR as no effect. This is despite, in the main, including the same studies (Supplementary Table S1). It should be noted that no published meta-analysis (or individual study) has shown a statistically significant reduction in kidney function in response to exercise training as compared to usual care, i.e., there is no detrimental effect. The results of the individual studies included in the systematic reviews, along with other relevant studies, are presented in Supplementary Table S2. Here, we critically analyse the published systematic reviews to summarise the current state of play in this field of research, identifying the reasons for the disparate findings, in order to suggest future research directions.
One of the key reasons for the difference in results is the choice of which eGFR outcome is used in each meta-analysis. Some compare the eGFR at final follow-up between exercise and usual care groups; others compare the change in eGFR from baseline. Clearly, the analyses that only compare final eGFR do not take into account differences in eGFR at baseline; this is an issue for small studies where randomisation may not result in similar values between groups. This is certainly the case in Greenwood et al. [10]: the baseline eGFR (CKD-EPIcr) was 36.6 in the exercise training group, compared to 46.5 in the controls. A similar discrepancy was seen in Headley et al. [13] (33.2 vs. 48.5); in other studies, the difference between exercise and control at baseline was 4 mL/min/1.73 m2 or less. Whilst these differences should be addressed by the meta-analysis methodology, where individual study results are combined and potential discrepancies in the randomisation are addressed by the increased total sample size, it remains possible that the groups were not equal in eGFR at baseline. It is also worth noting that only one of the included studies [10], gives information about the rates of decline prior to the studies, which may also be different between treatment groups, as it was in that study; the pre-intervention rate of decline may affect the response to treatment.
Nearly all the reviews include multiple studies with overlapping participant samples. Both studies by Aoike et al. and those by Baria and Gomes et al. were all sub-populations of the same study; specifically, Aoike et al., 2015 [104] was a 12-week duration and only the home-based exercise training group; Aoike et al., 2018 [112] was a 24-week duration, Baria et al., 2014 [102] reported males only and Gomes et al., 2017 [108] was a complete study similar to Aoike et al., 2018 [112], but with the two exercise groups (home and centre) combined. Similarly, Howden et al., 2013 and 2015 [8,120] report the same sub-population of which Beetham et al., 2018 [114] had a larger sample. Thus, all the systematic reviews, except Nakamura et al., 2020 [116] were biased by including results from the same patients multiple times.
There are other differences in the studies included in these reviews, which may also contribute to the difference in meta-analysis results. In the main, this appears to be due to differences in study inclusion criteria (for example, [113] is not included as it only recruited hypertensive non-diabetics; Ref. [12] had a factorial design that included dietary intervention) rather than a failure in systematic review methodology or conduct. An exception is Wu et al. [117]. Despite the methodology specifying randomised and quasi-randomised controlled trials, they included two single-arm studies [105,106], four randomised controlled trials [10,107,109,113] plus three studies only available in Chinese [121,122,123]; notably, these three studies were not included in Nakamura et al., 2020 [116] or Zhang et al., 2019 [118] despite both these reviews including Chinese language articles and searching Chinese databases. Similarly, Zhang et al., 2019 [118] includes [98], which is not randomised.
Table 1. Summary of meta-analyses of effect of exercise on kidney function.
Table 1. Summary of meta-analyses of effect of exercise on kidney function.
Review ReferenceExercise TrainingInclusionsStudies (or Strata) (n)Total Exercise ParticipantseGFR Results AnalysedeGFR Difference (mL/min/1.73 m2)
Yamamoto 2021 [115]Aerobic (± resistance)(e)GFR < 60
RCTs
1 month + duration
English language
Cardiometabolic or renal outcomes
11221Between-group mean final eGFR0.04
Nakamura 2020 [116]Aerobic or resistance or both(e)GFR < 60
RCTs or cross-over
1 month + duration
1+ per week
Intesity described
No language restriction
eGFR as outcome
10238Mixture of final eGFR and within-group difference in eGFR−0.34
Wu 2020 [117]Combined aerobic and resistanceCKD 1–5 (non-dialysis)
RCTs and quasi-RCTs
Any combined exercise intervention
Chinese or English language
eGFR as outcome
6143Between-group mean final eGFR a5.01 *
Villanego 2020 [17]Aerobic (± resistance)CKD 1–5 (non-dialysis)
assignment “described as random”
12 weeks + duration
English or Spanish language
13226Within-group change, intervention minus control−0.14
Zhang 2019 [118]Aerobic (± resistance)CKD 2–5 (non-dialysis)
RCTs
1 month + duration
1 + per week
Chinese or English language
eGFR as outcome
18262Within-group change, intervention minus control2.62 *
Vanden Wyngaert  2018 [119]Aerobic (± resistance)CKD 3–4 (non-dialysis)
RCTs
3 month + duration
2 + per week
Dutch, English, German or French language
eGFR as outcome
10154Within-group change, intervention minus control2.16 *
Findings of eGFR differences in meta-analyses of studies of exercise in ND-CKD. Positive numbers indicate a benefit to exercise training in reducing eGFR decline as compared with usual care. Statistically significant results are marked with an asterix (*). a Wu et al., 2020 also reported, in 9 studies with 329 participants, a statistically significant within-group benefit of exercise on eGFR of 3.01 mL/min/1.73 m2.
Other relevant studies have been excluded from these reviews, including [15,96,99]. These all used measured GFR (by 51Cr-EDTA, 125I-iothalamate clearance and iohexol clearance, respectively) rather than eGFR, which was the outcome of interest in the systematic reviews. Castaneda et al., 2001 [99] may also have been excluded because it was a trial of resistance-only exercise training, i.e., no aerobic component; also, Ref. [15] compared two different forms of exercise training without a usual care control. In addition, a Chinese study [103] comparing Tai Chi to usual care in the non-dialysis CKD population was not included in any systematic review. This study showed a statistically significant benefit on eGFR (an increase of 6 mL/min/m2 in the Tai Chi group). Presumably, this study was not included because Tai Chi is not widely considered as a form of exercise training, though it has been shown to have benefits on cardiorespiratory function, muscle strength and endothelial function [124,125].
Notwithstanding that the systematic reviews show contradictory results, there are other reasons to interpret their findings with caution. There is considerable heterogeneity in the study designs and the exercise interventions used, such that judgment about the generalisability of the findings is difficult. Additionally, the differences in study design reduce the legitimacy of combining the results by meta-analysis. Details of the included studies (and other studies with relevant outcomes) are found in Table 2. Firstly, there are differences in participant selection: whilst all selected patients with either CKD 2–4, 3 or 3–4, many studies specified other inclusion criteria, including requiring non-diabetic, type-2 diabetes mellitus (T2DM), hypertensive or obese patients or the presence of other cardiovascular risk factors. Secondly, there were important differences in the exercise interventions themselves. Some studies included light aerobic exercise only, including walking, for all or part of the programme [100,102,104,108,109,112]. Most others included a moderate aerobic component, but this was variably defined and prescribed: some studies used percentage of heart rate achieved during a maximal exercise tolerance test, others used the participant’s own Rating of Perceived Exertion (RPE), a subjective measure of the intensity of the activity. One study used short bouts of high-intensity cycling training [11]. Some studies also incorporated non-exercise elements in the interventions, which were not offered to the control groups. These included dietary advice or prescription [8,13,107,114,120]. The duration of interventions ranged from 12 to 52 weeks, with just one lasting longer at 156 weeks [110]; notably, this study demonstrated significant improvements in eGFR in both exercise groups at the end of the intervention, though there was no non-exercise control to compare with.
As well as noting the marked variability in intervention duration, it is worth considering whether these time frames are sufficient to see a difference in eGFR between treatment groups. The typical rate of decline in people with established CKD can vary from as little 2.4 to 8.5 mL/min/1.72 m2 per year [126]; the KDIGO international guideline defines >5 mL/min/1.72 m2 per year reduction in eGFR as rapid decline. Furthermore, it is widely recognised that decline in eGFR is not linear; most patients with CKD demonstrate considerable variability in their disease course, often with a prolonged period of non-progression [1,127]. Furthermore, one might compare the length of follow-up required in landmark studies of pharmacological interventions. Follow-up durations of 2.4 to 3.4 years have been needed to reach composite primary end-points, which included doubling of serum-creatinine and progression to ESKD (markers of CKD progression), in studies of key agents, such as angiotensin-converting enzyme inhibitors (ACEi) angiotensin receptor blockers (ARB) and SGLT-2 inhibitors [128,129,130,131,132,133].
Table 2. Characteristics of exercise training studies in non-dialysis CKD, with renal function as an outcome.
Table 2. Characteristics of exercise training studies in non-dialysis CKD, with renal function as an outcome.
Study ReferenceGroups (n)Study Duration (Weeks)Mean eGFR (Baseline)Other Population Inclusion CriteriaAge (Years)Exercise Frequency and TimeAerobic ExerciseResistance Exercise
Aoike 2015, 2018, Baria 2014, Gomes 2017 [102,104,108,112]H-ET (14);
C-ET (13);
UC (15)
12–2426.7BMI > 25; male only in Baria 20145530–50 min, 3 × per wkMild-Moderate (VT i.e., 40–60% VO2max): home-based = walking;
centre-based = treadmill
Nil
Barcellos 2018 [113]ET (76);
UC (74)
1669.0Hypertensive, non-diabetic6560 min, 3 × per wk, supervisedStep aerobic workout in 30 s bouts as part of circuit trainingBody weight, core and dumbbell exercises in 30 s bouts as part of circuit training
Beetham 2019 [7]HIIT (9);
Moderate ET (5)
1261.61+ uncontrolled CVD risk factors (BP, HbA1C, lipids)61HIIT: 4 × 4 min;
Moderate Ex: 40 min 3 days per wk
HIIT: 80–95% PeakHR, Mod Ex: 65% peakHR;
treadmill
Nil
Castaneda 2001/2004 [99,134]ET + low-protein diet (14);
low-protein diet only (12)
1229.5Age > 506445 min, 3 × per wkNil3 sets × 8 reps, upper and lower body exercises using resistance training machines; 80% of 1RM
Eidemak 1997 [96]ET (15);
UC (15)
7825Non-diabetic4430 min, daily;
unsupervised
60–75% maximal exercise capacity, static bike, running, swimming or walkingNil
Greenwood 2015 [10]ET (8);
UC (10)
5242.1 5340 min, 3 × per wk (2 × supervised, 1 × home)stationary exercise cycle;
80% HRR
3 sets × 8–10 reps, upper and lower body exercises using free weights or resistance bands; 80% of 1RM
Gregory 2011; Headley 2012 [13,135]ET (10);
UC (11)
4841.2 5545 min, 3 × per wk, supervised50–60% VO2peakWeeks 24–48;
1–3 sets × 10–12 reps; upper and lower body using weight machines
Hamada 2016 [106]ET (47)2647.7 6990–120 min, 6 × per monthWalking, RPE 12–143–4 METs
Headley 2014, 2017, Miele 2017 [111,136,137]ET (25);
UC (21)
1647.6DM or HTN as primary cause of CKD5830–45 min, 3 × per wk, supervised50–60% VO2peak, mixed apparatusNil
Hellberg 2019, Zhou 2020 [15,138]Endurance & strength (73);
Endurance and balance (75)
5219.5 66150 min/ wk2 × 30 min;
RPE 13–15
3 × 30 min; RPE 13–17;
2–3 sets × 10 reps; free weight/body weight (resistance) or balance exercises
Hiraki 2017 [109]H-ET (14);
UC (14)
5239.5Male6830 min daily or 8000–10,000 steps, plus resistanceWalkingHand grip, squats and calf raises;
20–30 reps 3× per week
Howden 2013,2015, Beetham 2018, Small 2017, Huppertz 2020 [8,114,120,139,140]ET and lifestyle intervention (81);
UC (80)
5239.61 + uncontrolled CVD risk factors (BP, HbA1C, lipids)62150 min per wk, beginning with 8 wks 2–3 × per wk supervised in gym;
subsequently at home
Moderate, RPE 11–13Whole-body with therabands and Swiss ball
Ikizler 2018 [12]ET vs usual activity;
Calorie restriction vs. usual diet (104 total)
1642.4BMI > 255730–45 min, 3 × per wk, supervised60–80% VO2peak;
cycling, treadmill or epliptical
Nil
Kirkman 2019 [141]ET (15);
UC (16)
1243 5845 min, 3 × per wk;
supervised
60–85% heart rate reserve;
cycling, treadmill or epliptical
Nil
Kiuchi 2017 [110]HIIT (25);
Mod ET (25)
15643.1HTN58HIIT: 4–30 min;
Mod Ex: 30–60 min 5 days per wk
HIIT: up to maximal intensity;
Mod Ex: 55–85% max HR
Nil
Leehey 2016 [107]ET + diet (14);
Diet alone (18)
5239.9T2DM, obese, male6660 min, 3 × per wk;
superveised for first 12 wks
45–85% VO2peak;
Treadmill, elliptical or cycle;
intervals
Additional 20–30 min/ session
Leehey 2009 [100]ET (7);
UC (4)
2445Diabetic, BMI > 30, proteinuria6630–40 min, 3 × per wk, supervised for first 6 wksWalking;
supervised sessions were up to 60–84% VO2peak
Nil
Mustata 2011 [101]ET (10);
UC (10)
5227.5 6820–60 min, 5 × per wk (3 unsupervised, 2 supervised)40–60% VO2peak, treadmill, cycle, ellipitcal trainer;
unsupervised = walking
Nil
Nylen 2015 [105]ET (128)1276.1T2DM6260 min, 2 × per weekMixed aerobic and resistance; 50–85% HRRNo details
Pechter 2003 [142]ET (17);
UC (9)
1265.3Proteinuria;
2 + CVD risk factors
4930 min, 2 × per wkAquatic exerciseNil
Shi 2014 [103]ET (11);
UC (10)
1245Evidence of CVD6930 min, 3–5 × per wk, supervised for first 4 wksTai ChiNil
Toyama 2010 [98]ET (10);
UC (9)
1247.4CVD7230 min, 7 × per wk (1 × supervised, 6 × home)1 × supervised bicycle ergometer;
daily walking, RPE 12–13
Nil
Van Craenenbroeck 2015 [11]ET (19);
UC (21)
1238.6 5310 min, 4 × per day, at homeStatic cycling, 90% HR of anaerobic thresholdNil
Abbreviations: BMI, body mass index; C-ET, centre-based exercise training; CVD, cardiovascular disease; DM, diabetes mellitus; ET, exercise training; H-ET, home-based exercise training; HIIT, high intensity interval training; HR, heart rate; HRR, heart rate reserve; HTN, hypertension; RPE, rating of perceived exertion; T2DM, Type 2 diabetes mellitus; UC, ususal care; VT, ventilatory threshold. Trials of exercise training in people with non-dialysis CKD, which include a measure of kidney function as an outcome. These studies are either included in recent meta-analyses or otherwised referenced in this review. Where multiple publications have been made from the same study (i.e., interim analyses or sub-samples), information for sample sizes, mean eGFR and age are taken from the most complete publication. All studies in the table were randomised-controlled trials except the following: Pechter 2003 and Toyama 2010 (non-randomised controlled trials); Hamada 2016 and Nylen 2015 (single-arm).
Mention should be made of the measures of kidney function used in these studies. As already alluded to, none of the included studies used directly measured GFR. The majority of studies included in the meta-analyses reviewed here use creatinine-based estimates of GFR with a variety of formulae (Supplementary Table S2). The influence of muscle mass and diet on serum creatinine concentrations [143] is of concern in exercise interventions where there is an expectation that muscle mass may increase. Cystatin C has been proposed as an alternative molecule that could be used for estimating GFR. Cystatin C is produced by all human nucleated cells and completely filtered at the glomerulus before being reabsorbed and metabolised by the tubular epithelium [144]. The use of cystatin C for estimating GFR is not without its own limitations due to some extra-renal elimination and influence of factors such as body mass, CRP and white blood cell count [145,146]. Cystatin C, however, appears to be less influenced by muscle mass [146,147], and so its use in exercise studies is preferable to creatinine-based estimates alone, although it can still lead to misinterpretation of kidney function in patients with very high or very low muscle mass [148]. Cystatin C-based measures were used in only two studies [12,114] (and as an exploratory analysis in [10]).
A key reason why studies may have failed to find a positive effect of exercise training, and another reason why it is difficult to draw conclusions on efficacy from the findings of the meta-analyses to date, is the issue of compliance. Compliance with an exercise programme, both in terms of attending exercise sessions and fidelity to the prescribed duration and intensity, is clearly important to assessing the efficacy of the training. These elements were another source of variability in the studies included in previous meta-analyses (if they were reported at all). The details of participant compliance are summarised in Table 3. Overall, where it is reported, compliance to session attendance was 70–97%, although it is likely to have been lower in some studies, including [12] (85% attended at least 50% of sessions) and [113] (63.8% attended at least 70% of sessions). Conversely, adherence/fidelity to the prescribed programme, i.e., to what extent—in terms of mode, intensity and time—did participants complete the prescribed exercise, is rarely reported. Study authors report using a variety of means to monitor the completion of prescribed exercise, including monitoring by completion of diaries. Heart rate monitors, which can give more accurate information about the duration and intensity of the exercise (when data is combined with that from a graded exercise tolerance test), were used in only a few studies and mainly without reporting of data (see Table 3). The baseline and follow-up testing of the VO2peak acts as a manipulation check to assess if interventions have occurred and been adhered to sufficiently to induce physiological change. Six studies showed improvements in cardiorespiratory fitness [8,10,11,13,111,112]; thus, even though we do not know much about the fidelity of exercise programmes, it was sufficient for some physiological adaptation. In contrast, however, an equal number of studies did not include a measure of cardiorespiratory fitness [109,110,113] or reported no change [12,100,107] in this outcome measure.
This leads to an important question: even if all ET programmes were designed to induce improvements in cardiorespiratory fitness, and the programmes were sufficiently adhered to gain these benefits, is this the same level of intensity, frequency or duration of activity sufficient to induce physiological changes that benefit kidney function? Perhaps a more fundamental question is what do we expect exercise training to do, physiologically or immunologically or otherwise, that would delay kidney disease progression? This will be considered in the following sections.

5. CKD Risk Factors Are Amenable to Exercise Interventions

There are a number of possible ways in which exercise may help delay CKD progression. Importantly, the CKD risk factors discussed above have been shown to be amenable to exercise interventions. The benefit of exercise training has been demonstrated in other disease populations, including improved blood pressure in elderly hypertensives and other groups [149,150]; improved glycaemic control in T2DM [151]; improved insulin sensitivity in T2DM and other populations [152,153]; and weight loss in overweight and obese adults [154].
There is also evidence that some of these risk factors can be successfully targeted with exercise training in patients with CKD. Evidence includes a number of systematic reviews with meta-analyses demonstrating positive effects on blood pressure and BMI [17,115,118,119,155,156], summarised in Table 4 and Table 5. Though not all statistically significant, a clinically meaningful mean reduction in systolic blood pressure (SBP) has been found in all these meta-analyses. The exception is Van den Wyngaert et al., 2018, though that review also noted a significant within-group reduction in SBP of 5.2 mmHg following exercise training. Again, the reviews analysing the effects on BMI find mean differences in favour of exercise training; in the main, these results are statistically significant. The effects of exercise training on other factors in CKD patients have been less well studied. The effect on glycaemic control has been reviewed, and no effect was found on HbA1C in one meta-analysis, which included seven studies and sub-strata [115]; mixed results were found in another systematic review [157].
Whilst meta-analyses have found positive effects on both BMI and BP in exercise studies in non-dialysis CKD, these findings do not consistently correlate with improved eGFR outcomes: e.g., Ref. [119] found a positive effect on eGFR, with no effect on BP, whilst [115] found positive effects on BMI and BP but not eGFR. There is a similar mismatch in the individual studies reviewed in these meta-analyses.

6. Potential Benefits of Exercise on the Pathological Processes of CKD Progression

We have seen above that exercise training in CKD patients produces physiological adaptations that produce improved cardiorespiratory fitness and muscle strength. It is reasonable to consider whether similar training may induce adaptations in the kidney.
Whilst the precise mechanisms have not been elucidated, the stimulants for exercise-induced physiological adaptations occur primarily due to muscle stretching. Muscle activity results in changes including intra-muscular energy consumption, intracellular pH and Ca2+ concentration, as well as the generation of mechanical forces by muscle contraction [158]. These stimulants have all been linked to changes in protein and enzyme transcription, which result in metabolic, hormonal and vascular changes, which then produce beneficial adaptations to exercise training. There are also stimulants that occur outside of the contracting muscles, including increased shear stress driven by increased cardiac output and blood flow and altered sympathetic nervous activity [159]. Clearly, physical stretching is not relevant to kidney tissue. Furthermore, during exercise, renal blood flow diminishes rather than increases. Hence, mechanisms specific to muscles (related to mechanical stretching and increased shear stress) are not replicated in the kidney. However, whilst these stimulants derive from active muscle tissue, the downstream effectors are not necessarily limited to muscles, and some are of relevance to renal pathophysiology. For example, skeletal muscle capillary growth is stimulated by mechanical influences, such as shear stress induced by increased blood flow, and stretching of the muscle tissue, with VEGF mediating the effect [160,161]. VEGF may have systemic and beneficial effects on maintaining the renal vasculature, particularly the glomerular and tubular capillaries, which are so relevant to the pathogenesis of CKD progression. There are a number of lines of evidence to suggest that the physiological changes that occur during exercise not only occur in the active skeletal muscle but also take place elsewhere in the body. Examples include an improvement in endothelial-dependent dilation in the brachial artery as a result of leg cycling training [162]. Distal vascular changes have also been demonstrated in studies where there is no increased cardiac load: in a four-week passive leg movement programme, there was an increased capillary density and VEGF concentration in the muscle of the untrained leg [163].
Another key adaptation to exercise training is mitochondrial biogenesis [164]. This is an important development, in response to Ca2+ cycling and ROS production etc., for improving the energy supply to active muscle. It would also be a useful adaptation in metabolically active kidney tissue, helping to optimise the use of oxygen to produce energy, potentially alleviating the effects of hypoxia in CKD pathophysiology. In animal models of diabetic and hypertensive nephropathy, aerobic exercise training produced beneficial effects on renal mitochondria, including inducing key enzymes and transcription factors for mitochondrial biogenesis, increasing mitochondrial ATP and reducing mitochondrial ROS production. These changes were associated with reduced renal disease progression compared with untrained animals [165,166,167].
Studies in humans have demonstrated reduced concentrations of some of the key molecules implicated in renal pathophysiology (discussed above) in response to exercise. These include MCP-1 [168,169], RANTES [170], NF- κ B [171] and TNF α [172]. Exercise also increases anti-oxidants and decreases pro-oxidants in a variety of healthy and chronic disease populations [173]. Alterations to levels of myokines, i.e., cytokines produced by active skeletal muscle tissue, may also be relevant because of their anti-inflammatory and metabolic effects. These include the effects of IL-6 on increasing fatty acid oxidation, thus increasing energy availability, and reducing TNF α production from macrophages, amongst other anti-inflammatory effects [174]. Another example is the myokine irisin, serum levels of which are decreased in patients with CKD [175,176] and can be increased with exercise training, e.g., in older adults [177]. Interestingly, a mouse model of CKD (interstitial fibrosis) demonstrated improved kidney mitochondrial energy metabolism and reduced fibrosis after irisin induction [178]. Whilst some of these changes have not yet been demonstrated in patients with CKD, exercise may help readjust the imbalance of these important mediators in this population.
Benefits on other inflammatory and oxidative stress molecules have been seen in patients with CKD: 12 weeks of exercise in CKD 3–4 ameliorated cutaneous microvascular endothelial dysfunction by reducing oxidative stress. This suggests systemic anti-oxidant and endothelial function benefits of exercise [141]. Similarly, exercise training in CKD has been shown to reduce serum markers of oxidative stress, including F2-isoprostane [12] and reduce lipid peroxidation and glutathione oxidation after 12 weeks of aquatic exercise in participants with mild-to-moderate CKD [142]. There is reduced (sodium oxide dismutase) SOD and Nrf2, important anti-oxidant/anti-inflammatory molecules in moderate CKD, compared with healthy controls; this was improved after acute exercise [179]. Likewise, resistance exercise in haemodialysis patients increases Nrf2 [180]. These are systemic anti-oxidant effects that may benefit ROS imbalance present in the diseased kidney.
It is important to consider the extent to which improved systemic concentrations of circulating molecules are reflective of intra-renal levels: i.e., does a systemic reduction in, for example, anti-oxidants after exercise translate into an improvement in oxidative stress in the glomerulus or tubules of the kidney? Such questions would require further animal studies to develop our understanding in this area.
Finally, whilst a bout of exercise leads to increased SNS activity [181], exercise training is known to dampen this response and may even reduce resting sympathetic tone [182]. Indeed, exercise training in healthy adults has been shown to result in reduced resting renal sympathetic activity, specifically [183]. As discussed, CKD patients have SNS overactivity, which may contribute to the pathophysiology of the disease. Thus, reduced SNS activity may be another way in which exercise training may affect CKD progression.
In summary, there are a number of potential ways in which exercise may reduce CKD progression. Prominent risk factors for CKD—BP and BMI—are improved by exercise, both in CKD patients and other populations. It is also plausible that exercise may affect the mediators of kidney disease pathophysiology themselves, with human exercise studies showing modification of many of the relevant molecules and pathways.

7. Summary and Discussion of Future Directions

Overall, the story so far is that, despite benefits on disease progression risk factors, the effect of exercise on kidney function itself is unclear. The heterogeneous findings of recent meta-analyses reflect the heterogeneity of the method of analysis and the studies included. Alongside this, in preventing a definitive conclusion, is the heterogeneity of individual study designs, particularly in terms of duration and intensity of exercise interventions. Uncertainty about participants’ compliance to the exercise programme also makes assessment of the efficacy difficult.
We have seen that it is probably unreasonable, given the typical rates and patterns of eGFR decline, to expect to see significant or meaningful differences after durations of exercise training less than 12 months. Recently, consideration has been given to using the slope of eGFR decline as an outcome in clinical trials [184,185,186] with a suggestion that slope over 2–3 years is a useful surrogate marker for more definitive end-points, such as reaching ESKD. We might also consider whether it is realistic to expect to meaningfully delay the rate of kidney function decline in patients with already advanced disease. More advanced CKD stage is consistently shown to be a risk factor for more rapid eGFR decline [60,84,187,188]. This is likely to be one reason why pharmacological intervention studies, such as those discussed herein recruit patients with a wider range of eGFR and often exclude those with more advanced CKD. Those designing future studies in this field need to consider who is likely to benefit: if patients are already at an advanced stage with progressive disease, is it too late to intervene? Intervention in earlier stages of CKD could be considered, but this would increase the chance of intervening on patients at low risk of decline who would not progress anyway. An alternative may be to select those at earlier stages of CKD with risk factors for kidney function decline.
As discussed, neither creatinine or cystatin C provide perfect estimates of GFR. Hence, consideration might be given to using directly measured GFR. Measuring GFR using exogenous filtration markers—such as 51Cr-EDTA, 125I-iothalamate clearance and iohexol clearance—has a strong rationale for studies specifically investigating effects on kidney function. These measures, however, are not without their own limitations, including a degree of intra-test and inter-test variability in measurements, more prominent in people with higher GFR [143]. Perhaps more critical is the practicality and patient acceptability of these tests, which may only be available in specialist centres and, in most cases, take a number of hours and multiple venepunctures to perform. In our experience, there has been some patient reluctance because of the travel and time involved. A recently updated GFR estimating formula, using both serum creatinine and cystatin C measurements, has shown good accuracy with a median difference between measured GFR and eGFR of 0.1 and −2.9 mL/min/1.73 m2 in black and non-black participants, respectively, and 91% of all estimates being within 30% of measured GFR [189]; accuracy was greater with lower GFR. In our opinion, eGFR using cystatin C in combination with creatinine is a practical and sufficiently accurate alternative to measured GFR for use in exercise studies, particularly in patients with eGFR < 60 and particularly if repeated measures of eGFR are available to calculate a slope of decline.
A large proportion of the studies included in the meta-analyses discussed here are relatively small and not designed to have power to detect differences in eGFR and are at risk of unequal groups at baseline. Therefore, as well as considering increased length of intervention, studies with larger sample sizes are needed to allow more robust randomisation; this is an important priority for future research. Multi-centre collaboration should be considered as a means to increasing sample size in studies whose primary outcome is change in kidney function.
Multiple barriers to exercise participation exist in patients living with CKD [190,191], so compliance is likely to always be an issue. This complicates assessing the efficacy of prescribed programmes, though this may reflect the effectiveness of exercise training in the real-world. Nevertheless, thought needs to be given to improving motivation to and accessibility of exercise training programmes to aid increased compliance, ideally without compromising on the physiological load so that studies continue to address the underlying scientific question. The mechanisms involved in exercise effects on kidney function remain to be seen, and this, too, deserves further attention. Our opinion is that the highest priority for future research into the effects of exercise on CKD progression should be a more targeted approach to patient selection, i.e., identifying those patients who are most likely to benefit. This entails (1) identifying patients who are at risk of disease progression, and (2) targeting populations in which the adaptations induced by exercise training are likely to be most impactful; this requires a better understanding of the mechanisms of kidney adaptation to exercise.
Current recommendations are that patients with ND-CKD should complete 150 min of moderate aerobic exercise per week plus resistance and flexibility exercise on at least 2 days per week [192]. Whilst it is not clear which, if any, of these forms of exercise are more likely to lead to improvements in kidney function decline, we believe future studies in this field should incorporate both aerobic and resistance exercise into their programmes for several reasons. Firstly, studies that have shown benefits on CKD risk factors have invariably included aerobic, and often resistance, exercise. Secondly, some of the mechanisms described above for altering inflammatory levels are dependent on muscle stretch or have been demonstrated in resistance-only programmes. Thus, resistance training should also be included. Finally, both of these forms of exercise have demonstrated benefits to CKD unrelated to kidney function and it might be considered inappropriate to omit one or the other.
There is uncertainty about the benefits of exercise on disease progression in CKD. Whilst no RCT to date has shown a detrimental effect, it would be inappropriate at this stage to commit to exercise training for this purpose at the level of service commissioning or national guideline recommendations. Despite this, as there is no evidence of harm from exercise intervention on kidney function, and considering the many other potential benefits of increased physical activity, exercise should be increasingly recommended and prescribed by health professionals. Future research, including cost effectiveness analyses may then provide sufficient evidence to support exercise training being delivered as part of routine care.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/kidneydial2020026/s1, Table S1: Studies included in published systematic reviews of effects of exercise on kidney function in ND-CKD. Table S2: Effect of Exercise Training on Kidney Function in NDD-CKD: summary of study results.

Author Contributions

Conceptualization, M.D., J.M. and A.S.; writing—original draft preparation, M.D.; writing—review and editing, M.D., J.M. and A.S. All authors have read and agreed to the published version of the manuscript.

Funding

Mark Davies’s PhD research is supported by Awyr Las, Registered Charity No. 1138976.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Li, L.; Astor, B.C.; Lewis, J.; Hu, B.; Appel, L.J.; Lipkowitz, M.S.; Toto, R.D.; Wang, X.; Wright, J.T.; Greene, T.H. Longitudinal progression trajectory of GFR among patients with CKD. Am. J. Kidney Dis. 2012, 59, 504–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tonelli, M.; Wiebe, N.; Culleton, B.; House, A.; Rabbat, C.; Fok, M.; McAlister, F.; Garg, A.X. Chronic Kidney Disease and mortality risk: A systematic review. J. Am. Soc. Nephrol. 2006, 17, 2034–2047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Go, A.S.; Chertow, G.M.; Fan, D.; McCulloch, C.E.; Hsu, C.Y. Chronic Kidney Disease and the Risks of Death, Cardiovascular Events, and Hospitalization. N. Engl. J. Med. 2004, 351, 1296–1305. [Google Scholar] [CrossRef] [PubMed]
  4. Mende, C.W. Chronic Kidney Disease and SGLT2 Inhibitors: A Review of the Evolving Treatment Landscape. Adv. Ther. 2022, 39, 148–164. [Google Scholar] [CrossRef]
  5. Shabaka, A.; Cases-Corona, C.; Fernandez-Juarez, G. Therapeutic Insights in Chronic Kidney Disease Progression. Front. Med. 2021, 8, 645187. [Google Scholar] [CrossRef]
  6. Yang, L.; Wu, X.; Wang, Y.; Wang, C.; Hu, R.; Wu, Y. Effects of exercise training on proteinuria in adult patients with Chronic Kidney Disease: A systematic review and meta-analysis. BMC Nephrol. 2020, 21, 172. [Google Scholar] [CrossRef]
  7. Beetham, K.S.; Howden, E.J.; Fassett, R.G.; Petersen, A.; Trewin, A.J.; Isbel, N.M.; Coombes, J.S. High-intensity interval training in Chronic Kidney Disease: A randomized pilot study. Scand. J. Med. Sci. Sports 2019, 29, 1197–1204. [Google Scholar] [CrossRef]
  8. Howden, E.J.; Coombes, J.S.; Strand, H.; Douglas, B.; Campbell, K.L.; Isbel, N.M. Exercise training in CKD: Efficacy, adherence, and safety. Am. J. Kidney Dis. 2015, 65, 583–591. [Google Scholar] [CrossRef]
  9. Rossi, A.P.; Burris, D.D.; Lucas, F.L.; Crocker, G.A.; Wasserman, J.C. Effects of a renal rehabilitation exercise program in patients with CKD: A randomized, controlled trial. Clin. J. Am. Soc. Nephrol. 2014, 9, 2052–2058. [Google Scholar] [CrossRef] [Green Version]
  10. Greenwood, S.A.; Koufaki, P.; Mercer, T.H.; MacLaughlin, H.L.; Rush, R.; Lindup, H.; O’Connor, E.; Jones, C.; Hendry, B.M.; Macdougall, I.C.; et al. Effect of Exercise Training on Estimated GFR, Vascular Health, and Cardiorespiratory Fitness in Patients With CKD: A Pilot Randomized Controlled Trial. Am. J. Kidney Dis. 2015, 65, 425–434. [Google Scholar] [CrossRef]
  11. Van Craenenbroeck, A.H.; Van Craenenbroeck, E.M.; Van Ackeren, K.; Vrints, C.J.; Conraads, V.M.; Verpooten, G.A.; Kouidi, E.; Couttenye, M.M. Effect of Moderate Aerobic Exercise Training on Endothelial Function and Arterial Stiffness in CKD Stages 3–4: A Randomized Controlled Trial. Am. J. Kidney Dis. 2015, 66, 285–296. [Google Scholar] [CrossRef] [PubMed]
  12. Ikizler, T.A.; Robinson-Cohen, C.; Ellis, C.; Headley, S.A.; Tuttle, K.; Wood, R.J.; Evans, E.E.; Milch, C.M.; Moody, K.A.; Germain, M.; et al. Metabolic effects of diet and exercise in patients with moderate to severe CKD: A randomized clinical trial. J. Am. Soc. Nephrol. 2018, 29, 250–259. [Google Scholar] [CrossRef] [PubMed]
  13. Headley, S.A.; Germain, M.; Milch, C.; Pescatello, L.; Coughlin, M.A.; Nindl, B.C.; Cornelius, A.; Sullivan, S.; Gregory, S.; Wood, R. Exercise training improves HR responses and VO2peak in predialysis kidney patients. Med. Sci. Sports Exerc. 2012, 44, 2392–2399. [Google Scholar] [CrossRef] [PubMed]
  14. Watson, E.L.; Gould, D.W.; Wilkinson, T.J.; Xenophontos, S.; Clarke, A.L.; Vogt, B.P.; Viana, J.L.; Smith, A.C. Twelve-week combined resistance and aerobic training confers greater benefits than aerobic training alone in nondialysis CKD. Am. J. Physiol. Ren. Physiol. 2018, 314, F1188–F1196. [Google Scholar] [CrossRef] [Green Version]
  15. Hellberg, M.; Höglund, P.; Svensson, P.; Clyne, N. Randomized Controlled Trial of Exercise in CKD-The RENEXC Study. Kidney Int. Rep. 2019, 4, 963–976. [Google Scholar] [CrossRef] [Green Version]
  16. Heiwe, S.; Jacobson, S.H. Exercise training in adults with CKD: A systematic review and meta-analysis. Am. J. Kidney Dis. 2014, 64, 383–393. [Google Scholar] [CrossRef]
  17. Villanego, F.; Naranjo, J.; Vigara, L.A.; Cazorla, J.M.; Montero, M.E.; García, T.; Torrado, J.; Mazuecos, A. Impact of physical exercise in patients with Chronic Kidney Disease: Sistematic review and meta-analysis. Nefrologia 2020, 40, 237–252. [Google Scholar] [CrossRef]
  18. Martins, P.; Marques, E.A.; Leal, D.V.; Ferreira, A.; Wilund, K.R.; Viana, J.L. Association between physical activity and mortality in end-stage kidney disease: A systematic review of observational studies. BMC Nephrol. 2021, 22, 227. [Google Scholar] [CrossRef]
  19. Clarke, A.L.; Zaccardi, F.; Gould, D.W.; Hull, K.L.; Smith, A.C.; Burton, J.O.; Yates, T. Association of self-reported physical function with survival in patients with Chronic Kidney Disease. Clin. Kidney J. 2019, 12, 122–128. [Google Scholar] [CrossRef]
  20. MacKinnon, H.J.; Wilkinson, T.J.; Clarke, A.L.; Gould, D.W.; O’Sullivan, T.F.; Xenophontos, S.; Watson, E.L.; Singh, S.J.; Smith, A.C. The association of physical function and physical activity with all-cause mortality and adverse clinical outcomes in nondialysis Chronic Kidney Disease: A systematic review. Ther. Adv. Chronic Dis. 2018, 9, 209. [Google Scholar] [CrossRef]
  21. Beddhu, S.; Wei, G.; Marcus, R.L.; Chonchol, M.; Greene, T. Light-Intensity Physical Activities and Mortality in the United States General Population and CKD Subpopulation. Clin. J. Am. Soc. Nephrol. 2015, 10, 1145–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wilkinson, T.J.; Clarke, A.L.; Nixon, D.G.D.; Hull, K.L.; Song, Y.; Burton, J.O.; Yates, T.; Smith, A.C. Prevalence and correlates of physical activity across kidney disease stages: An observational multicentre study. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. -Eur. Ren. Assoc. 2019, 36, 641–649. [Google Scholar] [CrossRef] [PubMed]
  23. Hawkins, M.S.; Sevick, M.A.; Richardson, C.R.; Fried, L.F.; Arena, V.C.; Kriska, A.M. Association between physical activity and kidney function: National health and nutrition examination survey. Med. Sci. Sports Exerc. 2011, 43, 1457–1464. [Google Scholar] [CrossRef] [PubMed]
  24. Robinson-Cohen, C.; Littman, A.J.; Duncan, G.E.; Roshanravan, B.; Alp Ikizler, T.; Himmelfarb, J.; Kestenbaum, B.R. Assessment of Physical Activity in Chronic Kidney Disease. J. Ren. Nutr. 2013, 23, 123–131. [Google Scholar] [CrossRef] [Green Version]
  25. Martens, R.J.H.; van der Berg, J.D.; Stehouwer, C.D.A.; Henry, R.M.A.; Bosma, H.; Dagnelie, P.C.; van Dongen, M.C.J.M.; Eussen, S.J.P.M.; Schram, M.T.; Sep, S.J.S.; et al. Amount and pattern of physical activity and sedentary behavior are associated with kidney function and kidney damage: The Maastricht Study. PLoS ONE 2018, 13, e0195306. [Google Scholar] [CrossRef]
  26. Guo, C.; Tam, T.; Bo, Y.; Chang, L.Y.Y.; Lao, X.Q.; Thomas, G.N.; Lao, Q. Habitual physical activity, renal function and Chronic Kidney Disease: A cohort study of nearly 200,000 adults. Br. J. Sports Med. 2020, 54, 1225–1230. [Google Scholar] [CrossRef]
  27. Robinson-Cohen, C.; Littman, A.J.; Duncan, G.E.; Weiss, N.S.; Sachs, M.C.; Ruzinski, J.; Kundzins, J.; Rock, D.; de Boer, I.H.; Alp Ikizler, T.; et al. Physical Activity and Change in Estimated GFR among Persons with CKD. J. Am. Soc. Nephrol. 2014, 25, 399–406. [Google Scholar] [CrossRef] [Green Version]
  28. Robinson-Cohen, C.; Katz, R.; Mozaffarian, D.; Dalrymple, L.S.; de Boer, I.; Sarnak, M.; Shlipak, M.; Siscovick, D.; Kestenbaum, B. Physical Activity and Rapid Decline in Kidney Function Among Older Adults. Arch. Intern. Med. 2009, 169, 2116. [Google Scholar] [CrossRef] [Green Version]
  29. Taal, M.W. Mechanisms of Progression in Chronic Kidney Disease. In Brenner and Rector’s the Kidney, 11th ed.; Yu, A., Chertow, G., Luyckx, V., Marsden, P., Skorecki, K., Taal, M., Eds.; Elsevier: Amsterdam, The Netherlands, 2020; Chapter 51; pp. 1742–1789. [Google Scholar]
  30. Agarwal, A.; Nath, K.A. Chapter 18—Pathophysiology of Chronic Kidney Disease Progression: Organ and Cellular Considerations. In Chronic Renal Disease, 2nd ed.; Kimmel, P.L., Rosenberg, M.E., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 263–278. [Google Scholar]
  31. Stenvinkel, P.; Chertow, G.M.; Devarajan, P.; Levin, A.; Andreoli, S.P.; Bangalore, S.; Warady, B.A. Chronic Inflammation in Chronic Kidney Disease Progression: Role of Nrf2. Kidney Int. Rep. 2021, 6, 1775–1787. [Google Scholar] [CrossRef]
  32. Fine, L.G.; Norman, J.T. Chronic hypoxia as a mechanism of progression of Chronic Kidney Diseases: From hypothesis to novel therapeutics. Kidney Int. 2008, 74, 867–872. [Google Scholar] [CrossRef] [Green Version]
  33. Kriz, W.; LeHir, M. Pathways to nephron loss starting from glomerular diseases-insights from animal models. Kidney Int. 2005, 67, 404–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Harris, R.C.; Neilson, E.G. Toward a unified theory of renal progression. Annu. Rev. Med. 2006, 57, 365–380. [Google Scholar] [CrossRef] [PubMed]
  35. Mimura, I.; Nangaku, M. The suffocating kidney: Tubulointerstitial hypoxia in end-stage renal disease. Nat. Rev. Nephrol. 2010, 6, 667–678. [Google Scholar] [CrossRef] [PubMed]
  36. Theilig, F. Spread of glomerular to tubulointerstitial disease with a focus on proteinuria. Ann. Anat. 2010, 192, 125–132. [Google Scholar] [CrossRef]
  37. Jefferson, J.A.; Alpers, C.E.; Shankland, S.J. Podocyte biology for the bedside. Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 2011, 58, 835–845. [Google Scholar] [CrossRef] [Green Version]
  38. Zoja, C.; Benigni, A.; Remuzzi, G. Cellular responses to protein overload: Key event in renal disease progression. Curr. Opin. Nephrol. Hypertens. 2004, 13, 31–37. [Google Scholar] [CrossRef]
  39. Brenner, B.M.; Meyer, T.W.; Hostetter, T.H. Dietary Protein Intake and the Progressive Nature of Kidney Disease. N. Engl. J. Med. 1982, 307, 652–659. [Google Scholar]
  40. Takahashi, T.; Harris, R.C. Role of Endothelial Nitric Oxide Synthase in Diabetic Nephropathy: Lessons from Diabetic eNOS Knockout Mice. J. Diabetes Res. 2014, 2014, 590541. [Google Scholar] [CrossRef] [Green Version]
  41. Kida, Y.; Tchao, B.N.; Yamaguchi, I. Peritubular capillary rarefaction: A new therapeutic target in Chronic Kidney Disease. Pediatr. Nephrol. 2014, 29, 333–342. [Google Scholar] [CrossRef] [Green Version]
  42. Norman, J.T.; Fine, L.G. Intrarenal oxygenation in chronic renal failure. Clin. Exp. Pharmacol. Physiol. 2006, 33, 989–996. [Google Scholar] [CrossRef]
  43. Liu, Y. Cellular and molecular mechanisms of renal fibrosis. Nat. Rev. Nephrol. 2011, 7, 684–696. [Google Scholar] [CrossRef] [PubMed]
  44. Kang, D.H.; Kanellis, J.; Hugo, C.; Truong, L.; Anderson, S.; Kerjaschki, D.; Schreiner, G.F.; Johnson, R.J. Role of the microvascular endothelium in progressive renal disease. J. Am. Soc. Nephrol. 2002, 13, 806–816. [Google Scholar] [CrossRef] [PubMed]
  45. Lin, S.L.; Chang, F.C.; Schrimpf, C.; Chen, Y.T.; Wu, C.F.; Wu, V.C.; Chiang, W.C.; Kuhnert, F.; Kuo, C.J.; Chen, Y.M.; et al. Targeting endothelium-pericyte cross talk by inhibiting VEGF receptor signaling attenuates kidney microvascular rarefaction and fibrosis. Am. J. Pathol. 2011, 178, 911–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Rüster, C.; Wolf, G. Renin-angiotensin-aldosterone system and progression of renal disease. J. Am. Soc. Nephrol. 2006, 17, 2985–2991. [Google Scholar] [CrossRef] [PubMed]
  47. Tirichen, H.; Yaigoub, H.; Xu, W.; Wu, C.; Li, R.; Li, Y. Mitochondrial Reactive Oxygen Species and Their Contribution in Chronic Kidney Disease Progression Through Oxidative Stress. Front. Physiol. 2021, 12, 627837. [Google Scholar] [CrossRef]
  48. Ratliff, B.B.; Abdulmahdi, W.; Pawar, R.; Wolin, M.S. Oxidant Mechanisms in Renal Injury and Disease. Antioxidants Redox Signal. 2016, 25, 119–146. [Google Scholar] [CrossRef] [Green Version]
  49. Zhang, H.; Sun, S.C. NF-κB in inflammation and renal diseases. Cell Biosci. 2015, 5, 63. [Google Scholar] [CrossRef] [Green Version]
  50. Akchurin, O.M.; Kaskel, F. Update on inflammation in Chronic Kidney Disease. Blood Purif. 2015, 39, 84–92. [Google Scholar] [CrossRef]
  51. Grande, M.T.; Pérez-Barriocanal, F.; López-Novoa, J.M. Role of inflammation in túbulo-interstitial damage associated to obstructive nephropathy. J. Inflamm. 2010, 7, 19. [Google Scholar] [CrossRef] [Green Version]
  52. Neumann, J.; Ligtenberg, G.; Klein, I.I.; Koomans, H.A.; Blankestijn, P.J. Sympathetic hyperactivity in Chronic Kidney Disease: Pathogenesis, clinical relevance, and treatment. Kidney Int. 2004, 65, 1568–1576. [Google Scholar] [CrossRef] [Green Version]
  53. Kaur, J.; Young, B.E.; Fadel, P.J. Sympathetic Overactivity in Chronic Kidney Disease: Consequences and Mechanisms. Int. J. Mol. Sci. 2017, 18, 1682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kiuchi, M.G.; Graciano, M.L.; Carreira, M.A.M.d.Q.; Kiuchi, T.; Chen, S.; Lugon, J.R. Long-Term Effects of Renal Sympathetic Denervation on Hypertensive Patients with Mild to Moderate Chronic Kidney Disease. J. Clin. Hypertens. 2016, 18, 190–196. [Google Scholar] [CrossRef] [PubMed]
  55. Johnson, R.J.; Schreiner, G.F. Hypothesis: The role of acquired tubulointerstitial disease in the pathogenesis of salt-dependent hypertension. Kidney Int. 1997, 52, 1169–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Klag, M.J.; Whelton, P.K.; Randall, B.L.; Neaton, J.D.; Brancati, F.L.; Ford, C.E.; Shulman, N.B.; Stamler, J. Blood Pressure and End-Stage Renal Disease in Men. N. Engl. J. Med. 2002, 334, 13–18. [Google Scholar] [CrossRef] [PubMed]
  57. Hallan, S.I.; Ritz, E.; Lydersen, S.; Romundstad, S.; Kvenild, K.; Orth, S.R. Combining GFR and Albuminuria to Classify CKD Improves Prediction of ESRD. J. Am. Soc. Nephrol. 2009, 20, 1069–1077. [Google Scholar] [CrossRef] [Green Version]
  58. Hsu, C.Y.; Iribarren, C.; McCulloch, C.E.; Darbinian, J.; Go, A.S. Risk Factors for End-Stage Renal Disease. Arch. Intern. Med. 2009, 169, 342. [Google Scholar] [CrossRef] [Green Version]
  59. Inaguma, D.; Imai, E.; Takeuchi, A.; Ohashi, Y.; Watanabe, T.; Nitta, K.; Akizawa, T.; Matsuo, S.; Makino, H.; Hishida, A. Risk factors for CKD progression in Japanese patients: Findings from the Chronic Kidney Disease Japan Cohort (CKD-JAC) study. Clin. Exp. Nephrol. 2017, 21, 446–456. [Google Scholar] [CrossRef] [Green Version]
  60. Hunsicker, L.; Adler, S.; Caggiula, A.; England, B.K.; Greene, T.; Kusek, J.W.; Rogers, N.L.; Teschan, P.E. Predictors of the progression of renal disease in the Modification of Diet in Renal Disease Study. Kidney Int. 1997, 51, 1908–1919. [Google Scholar]
  61. Levin, A.; Djurdjev, O.; Beaulieu, M.; Er, L. Variability and Risk Factors for Kidney Disease Progression and Death Following Attainment of Stage 4 CKD in a Referred Cohort. Am. J. Kidney Dis. 2008, 52, 661–671. [Google Scholar] [CrossRef]
  62. Appel, L.J.; Wright, J.T.J.; Greene, T.; Agodoa, L.Y.; Astor, B.C.; Bakris, G.L.; Cleveland, W.H.; Charleston, J.; Contreras, G.; Faulkner, M.L.; et al. Intensive blood-pressure control in hypertensive Chronic Kidney Disease. N. Engl. J. Med. 2010, 363, 918–929. [Google Scholar] [CrossRef] [Green Version]
  63. Ku, E.; Sarnak, M.; Toto, R.; McCulloch, C.E.; Lin, F.; Smogorzewski, M.; Hsu, C.Y. Effect of Blood Pressure Control on Long-Term Risk of End-Stage Renal Disease and Death Among Subgroups of Patients with Chronic Kidney Disease. J. Am. Heart Assoc. 2019, 8, e012749. [Google Scholar] [CrossRef] [PubMed]
  64. Sarafidis, P.A.; Ruilope, L.M.; Loutradis, C.; Gorostidi, M.; de la Sierra, A.; de la Cruz, J.J.; Vinyoles, E.; Divisón-Garrote, J.A.; Segura, J.; Banegas, J.R. Blood pressure variability increases with advancing Chronic Kidney Disease stage: A cross-sectional analysis of 16546 hypertensive patients. J. Hypertens. 2018, 36, 1076–1085. [Google Scholar] [CrossRef] [PubMed]
  65. Parati, G.; Ochoa, J.E.; Lombardi, C.; Bilo, G. Blood Pressure Variability: Assessment, Predictive Value, and Potential as a Therapeutic Target. Curr. Hypertens. Rep. 2015, 17, 23. [Google Scholar] [CrossRef] [PubMed]
  66. Jhee, J.H.; Seo, J.; Lee, C.J.; Park, J.T.; Han, S.H.; Kang, S.W.; Park, S.; Yoo, T.H. Ambulatory blood pressure variability and risk of cardiovascular events, all-cause mortality, and progression of kidney disease. J. Hypertens. 2020, 38, 1712–1721. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, Q.; Wang, Y.; Wang, J.; Zhang, L.; Zhao, M.H. Short-term systolic blood pressure variability and kidney disease progression in patients with Chronic Kidney Disease: Results from c-stride. J. Am. Heart Assoc. 2020, 9, e015359. [Google Scholar] [CrossRef]
  68. Carlström, M.; Wilcox, C.S.; Arendshorst, W.J. Renal autoregulation in health and disease. Physiol. Rev. 2015, 95, 405–511. [Google Scholar] [CrossRef] [Green Version]
  69. Bidani, A.K.; Griffin, K.A. Pathophysiology of hypertensive renal damage: Implications for therapy. Hypertension 2004, 44, 595–601. [Google Scholar] [CrossRef] [Green Version]
  70. Ravera, M.; Re, M.; Deferrari, L.; Vettoretti, S.; Deferrari, G. Importance of blood pressure control in Chronic Kidney Disease. J. Am. Soc. Nephrol. 2006, 17, S98–S103. [Google Scholar] [CrossRef]
  71. Sun, D.; Wang, J.J.; Wang, W.; Wang, J.; Wang, L.N.; Yao, L.; Sun, Y.H.; Li, Z.L. Human podocyte injury in the early course of hypertensive renal injury. World J. Clin. Cases 2019, 7, 3698–3710. [Google Scholar] [CrossRef]
  72. Dharmashankar, K.; Widlansky, M.E. Vascular endothelial function and hypertension: Insights and directions. Curr. Hypertens. Rep. 2010, 12, 448–455. [Google Scholar] [CrossRef] [Green Version]
  73. Palm, F.; Nordquist, L. Renal oxidative stress, oxygenation, and hypertension. Am. J. Physiol. -Regul. Integr. Comp. Physiol. 2011, 301, R1229–R1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Chien, K.L.; Lin, H.J.; Lee, B.C.; Hsu, H.C.; Lee, Y.T.; Chen, M.F. A prediction model for the risk of incident Chronic Kidney Disease. Am. J. Med. 2010, 123, 836–846. [Google Scholar] [CrossRef] [PubMed]
  75. Hsu, C.-Y.; McCulloch, C.E.; Iribarren, C.; Darbinian, J.; Go, A.S. Body mass index and risk for end-stage renal disease. Ann. Intern. Med. 2006, 144, 21–28. [Google Scholar] [CrossRef] [PubMed]
  76. Chang, A.R.; Grams, M.E.; Ballew, S.H.; Bilo, H.; Correa, A.; Evans, M.; Gutierrez, O.M.; Hosseinpanah, F.; Iseki, K.; Kenealy, T.; et al. Adiposity and risk of decline in glomerular filtration rate: Meta-analysis of individual participant data in a global consortium. BMJ 2019, 364, k5301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Navaneethan, S.D.; Yehnert, H.; Moustarah, F.; Schreiber, M.J.; Schauer, P.R.; Beddhu, S. Weight Loss Interventions in Chronic Kidney Disease: A Systematic Review and Meta-analysis. CJASN 2009, 4, 1565–1574. [Google Scholar] [CrossRef] [Green Version]
  78. Bolignano, D.; Zoccali, C. Effects of Weight Loss on Renal Function in Obese CKD Patients: A Systematic Review. Nephrol. Dial. Transplant. 2013, 28, iv82–iv98. [Google Scholar] [CrossRef] [Green Version]
  79. Bilha, S.C.; Nistor, I.; Nedelcu, A.; Kanbay, M.; Scripcariu, V.; Timofte, D.; Siriopol, D.; Covic, A. The Effects of Bariatric Surgery on Renal Outcomes: A Systematic Review and Meta-analysis. Obes. Surg. 2018, 28, 3815–3833. [Google Scholar] [CrossRef]
  80. Wolf, G. After All Those Fat Years: Renal Consequences of Obesity. Nephrol. Dial. Transplant. 2003, 18, 2471–2474. [Google Scholar] [CrossRef] [Green Version]
  81. Chagnac, A.; Weinstein, T.; Herman, M.; Hirsh, J.; Gafter, U.; Ori, Y. The effects of weight loss on renal function in patients with severe obesity. J. Am. Soc. Nephrol. 2003, 14, 1480–1486. [Google Scholar] [CrossRef] [Green Version]
  82. Couser, W.G.; Remuzzi, G.; Mendis, S.; Tonelli, M. The contribution of Chronic Kidney Disease to the global burden of major noncommunicable diseases. Kidney Int. 2011, 80, 1258–1270. [Google Scholar] [CrossRef] [Green Version]
  83. Haroun, M.K.; Jaar, B.G.; Hoffman, S.C.; Comstock, G.W.; Klag, M.J.; Coresh, J. Risk Factors for Chronic Kidney Disease: A Prospective Study of 23,534 Men and Women in Washington County, Maryland. J. Am. Soc. Nephrol. 2003, 14, 2934–2941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Tsai, C.W.; Ting, I.W.; Yeh, H.C.; Kuo, C.C. Longitudinal change in estimated GFR among CKD patients: A 10-year follow-up study of an integrated kidney disease care program in Taiwan. PLoS ONE 2017, 12, e0173843. [Google Scholar] [CrossRef] [PubMed]
  85. The Diabetes Control and Complications Trial Research Group. The Effect of Intensive Treatment of Diabetes on the Development and Progression of Long-Term Complications in Insulin-Dependent Diabetes Mellitus. N. Engl. J. Med. 1993, 29, 977–986. [Google Scholar]
  86. Zoungas, S.; Arima, H.; Gerstein, H.C.; Holman, R.R.; Woodward, M.; Reaven, P.; Hayward, R.A.; Craven, T.; Coleman, R.L.; Chalmers, J. Effects of intensive glucose control on microvascular outcomes in patients with type 2 diabetes: A meta-analysis of individual participant data from randomised controlled trials. Lancet Diabetes Endocrinol. 2017, 5, 431–437. [Google Scholar] [CrossRef]
  87. Schrijvers, B.F.; De Vriese, A.S.; Flyvbjerg, A. From hyperglycemia to diabetic kidney disease: The role of metabolic, hemodynamic, intracellular factors and growth factors/cytokines. Endocr. Rev. 2004, 25, 971–1010. [Google Scholar] [CrossRef]
  88. Reidy, K.; Kang, H.M.; Hostetter, T.; Susztak, K. Molecular mechanisms of diabetic kidney disease. J. Clin. Investig. 2014, 124, 2333–2340. [Google Scholar] [CrossRef]
  89. Giunti, S.; Barit, D.; Cooper, M.E. Mechanisms of diabetic nephropathy: Role of hypertension. Hypertension 2006, 48, 519–526. [Google Scholar] [CrossRef] [Green Version]
  90. Spoto, B.; Pisano, A.; Zoccali, C. Insulin resistance in Chronic Kidney Disease: A systematic review. Am. J. Physiol. Ren. Physiol. 2016, 311, F1087–F1108. [Google Scholar] [CrossRef] [Green Version]
  91. Rochlani, Y.; Pothineni, N.V.; Kovelamudi, S.; Mehta, J.L. Metabolic syndrome: Pathophysiology, management, and modulation by natural compounds. Ther. Adv. Cardiovasc. Dis. 2017, 11, 215–225. [Google Scholar] [CrossRef]
  92. Gallagher, E.J.; Leroith, D.; Karnieli, E. The metabolic syndrome—From insulin resistance to obesity and diabetes. Med. Clin. N. Am. 2011, 95, 855–873. [Google Scholar] [CrossRef]
  93. Alizadeh, S.; Ahmadi, M.; Ghorbani Nejad, B.; Djazayeri, A.; Shab-Bidar, S. Metabolic syndrome and its components are associated with increased Chronic Kidney Disease risk: Evidence from a meta-analysis on 11,109,003 participants from 66 studies. Int. J. Clin. Pract. 2018, 72, e13201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kurella, M.; Lo, J.C.; Chertow, G.M. Metabolic Syndrome and the Risk for Chronic Kidney Disease among Nondiabetic Adults. J. Am. Soc. Nephrol. 2005, 16, 2134–2140. [Google Scholar] [CrossRef] [PubMed]
  95. Whaley-Connell, A.; Sowers, J.R. Insulin Resistance in Kidney Disease: Is There a Distinct Role Separate from That of Diabetes or Obesity. Cardiorenal Med. 2017, 8, 41–49. [Google Scholar] [CrossRef] [PubMed]
  96. Eidemak, I.; Haaber, A.B.; Feldt-Rasmussen, B.; Kanstrup, I.L.L.; Strandgaard, S. Exercise training and the progression of chronic renal failure. Nephron 1997, 75, 36–40. [Google Scholar] [CrossRef] [PubMed]
  97. Pechter, Ü.; Ots, M.; Mesikepp, S.; Zilmer, K.; Kullissaar, T.; Vihalemm, T.; Zilmer, M.; Maaroos, J. Beneficial effects of water-based exercise in patients with Chronic Kidney Disease. Int. J. Rehabil. Res. 2003, 26, 153–156. [Google Scholar] [PubMed]
  98. Toyama, K.; Sugiyama, S.; Oka, H.; Sumida, H.; Ogawa, H. Exercise therapy correlates with improving renal function through modifying lipid metabolism in patients with cardiovascular disease and Chronic Kidney Disease. J. Cardiol. 2010, 56, 142–146. [Google Scholar] [CrossRef] [Green Version]
  99. Castaneda, C.; Gordon, P.L.; Uhlin, K.L.; Levey, A.S.; Kehayias, J.J.; Dwyer, J.T.; Fielding, R.A.; Roubenoff, R.; Singh, M.F. Resistance training to counteract the catabolism of a low-protein diet in patients with chronic renal insufficiency. A randomized, controlled trial. Ann. Intern. Med. 2001, 135, 965–976. [Google Scholar] [CrossRef]
  100. Leehey, D.J.; Moinuddin, I.; Bast, J.P.; Qureshi, S.; Jelinek, C.S.; Cooper, C.; Edwards, L.C.; Smith, B.M.; Collins, E.G. Aerobic exercise in obese diabetic patients with Chronic Kidney Disease: A randomized and controlled pilot study. Cardiovasc. Diabetol. 2009, 8, 62. [Google Scholar] [CrossRef] [Green Version]
  101. Mustata, S.; Groeneveld, S.; Davidson, W.; Ford, G.; Kiland, K.; Manns, B. Effects of exercise training on physical impairment, arterial stiffness and health-related quality of life in patients with Chronic Kidney Disease: A pilot study. Int. Urol. Nephrol. 2011, 43, 1133–1141. [Google Scholar] [CrossRef]
  102. Baria, F.; Kamimura, M.A.; Aoike, D.T.; Ammirati, A.; Leister Rocha, M.; de Mello, M.T.; Cuppari, L. Randomized controlled trial to evaluate the impact of aerobic exercise on visceral fat in overweight Chronic Kidney Disease patients. Nephrol. Dial. Transplant. 2014, 29, 857–864. [Google Scholar] [CrossRef]
  103. Shi, Z.M.; Wen, H.P.; Liu, F.R.; Yao, C.X. The effects of tai chi on the renal and cardiac functions of patients with Chronic Kidney and Cardiovascular Diseases. J. Phys. Ther. Sci. 2014, 26, 1733–1736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Aoike, D.T.; Baria, F.; Kamimura, M.A.; Ammirati, A.; de Mello, M.T.; Cuppari, L. Impact of home-based aerobic exercise on the physical capacity of overweight patients with Chronic Kidney Disease. Int. Urol. Nephrol. 2015, 47, 359–367. [Google Scholar] [CrossRef] [PubMed]
  105. Nylen, E.S.; Gandhi, S.M.; Kheirbek, R.; Kokkinos, P. Enhanced fitness and renal function in Type 2 diabetes. Diabet. Med. 2015, 32, 1342–1345. [Google Scholar] [CrossRef] [PubMed]
  106. Hamada, M.; Yasuda, Y.; Kato, S.; Arafuka, H.; Goto, M.; Hayashi, M.; Kajita, E.; Maruyama, S. The effectiveness and safety of modest exercise in Japanese patients with Chronic Kidney Disease: A single-armed interventional study. Clin. Exp. Nephrol. 2016, 20, 204–211. [Google Scholar] [CrossRef] [PubMed]
  107. Leehey, D.J.; Collins, E.; Kramer, H.J.; Cooper, C.; Butler, J.; McBurney, C.; Jelinek, C.; Reda, D.; Edwards, L.; Garabedian, A.; et al. Structured exercise in obese diabetic patients with Chronic Kidney Disease: A randomized controlled trial. Am. J. Nephrol. 2016, 44, 54–62. [Google Scholar] [CrossRef]
  108. Gomes, T.S.; Aoike, D.T.; Baria, F.; Graciolli, F.G.; Moyses, R.M.A.; Cuppari, L. Effect of Aerobic Exercise on Markers of Bone Metabolism of Overweight and Obese Patients with Chronic Kidney Disease. J. Ren. Nutr. 2017, 27, 364–371. [Google Scholar] [CrossRef]
  109. Hiraki, K.; Shibagaki, Y.; Izawa, K.P.; Hotta, C.; Wakamiya, A.; Sakurada, T.; Yasuda, T.; Kimura, K. Effects of home-based exercise on pre-dialysis Chronic Kidney Disease patients: A randomized pilot and feasibility trial. BMC Nephrol. 2017, 18, 198. [Google Scholar] [CrossRef] [Green Version]
  110. Kiuchi, M.G.; Chen, S.; Hoye, N.A. The effects of different physical activities on atrial fibrillation in patients with hypertension and Chronic Kidney Disease. Kidney Res. Clin. Pract. 2017, 36, 264–273. [Google Scholar] [CrossRef] [Green Version]
  111. Miele, E.M.; Headley, S.A.; Germain, M.; Joubert, J.; Herrick, S.; Milch, C.; Evans, E.; Cornelius, A.; Brewer, B.; Taylor, B.; et al. High-density lipoprotein particle pattern and overall lipid responses to a short-term moderate-intensity aerobic exercise training intervention in patients with Chronic Kidney Disease. Clin. Kidney J. 2017, 10, 524–531. [Google Scholar] [CrossRef]
  112. Aoike, D.T.; Baria, F.; Kamimura, M.A.; Ammirati, A.; Cuppari, L. Home-based versus center-based aerobic exercise on cardiopulmonary performance, physical function, quality of life and quality of sleep of overweight patients with Chronic Kidney Disease. Clin. Exp. Nephrol. 2018, 22, 87–98. [Google Scholar] [CrossRef]
  113. Barcellos, F.C.; Del Vecchio, F.B.; Reges, A.; Mielke, G.; Santos, I.S.; Umpierre, D.; Bohlke, M.; Hallal, P.C. Exercise in patients with hypertension and Chronic Kidney Disease: A randomized controlled trial. J. Hum. Hypertens. 2018, 32, 397–407. [Google Scholar] [CrossRef] [PubMed]
  114. Beetham, K.S.; Howden, E.J.; Krishnasamy, R.; Isbel, N.M.; Coombes, J.S. Feasibility of higher intensity exercise in patients with Chronic Kidney Disease. J. Sport. Med. Phys. Fit. 2018, 58, 127–134. [Google Scholar] [CrossRef] [PubMed]
  115. Yamamoto, R.; Ito, T.; Nagasawa, Y.; Matsui, K.; Egawa, M.; Nanami, M.; Isaka, Y.; Okada, H. Efficacy of aerobic exercise on the cardiometabolic and renal outcomes in patients with Chronic Kidney Disease: A systematic review of randomized controlled trials. J. Nephrol. 2021, 34, 155–164. [Google Scholar] [CrossRef]
  116. Nakamura, K.; Sasaki, T.; Yamamoto, S.; Hayashi, H.; Ako, S.; Tanaka, Y. Effects of exercise on kidney and physical function in patients with non-dialysis Chronic Kidney Disease: A systematic review and meta-analysis. Sci. Rep. 2020, 10, 18195. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, X.; Yang, L.; Wang, Y.; Wang, C.; Hu, R.; Wu, Y. Effects of combined aerobic and resistance exercise on renal function in adult patients with Chronic Kidney Disease: A systematic review and meta-analysis. Clin. Rehabil. 2020, 34, 851–865. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, L.; Wang, Y.; Xiong, L.; Luo, Y.; Huang, Z.; Yi, B. Exercise therapy improves eGFR, and reduces blood pressure and BMI in non-dialysis CKD patients: Evidence from a meta-analysis. BMC Nephrol. 2019, 20, 398. [Google Scholar] [CrossRef]
  119. Vanden Wyngaert, K.; Van Craenenbroeck, A.H.; Van Biesen, W.; Dhondt, A.; Tanghe, A.; Van Ginckel, A.; Celie, B.; Calders, P. The effects of aerobic exercise on eGFR, blood pressure and VO2peak in patients with Chronic Kidney Disease stages 3–4: A systematic review and meta-analysis. PLoS ONE 2018, 13, e0203662. [Google Scholar] [CrossRef]
  120. Howden, E.J.; Leano, R.; Petchey, W.; Coombes, J.S.; Isbel, N.M.; Marwick, T.H. Effects of Exercise and Lifestyle Intervention on Cardiovascular Function in CKD. Clin. J. Am. Soc. Nephrol. 2013, 8, 1494–1501. [Google Scholar] [CrossRef] [Green Version]
  121. Liang, F.; Chao, M.; Wang, Z.; Al, E. The effects of exercises of different types on renal function and the risk factors associated with cardiovascular disease in patients with Chronic Kidney Disease. Chin. J. Rehabil. Med. 2016, 31, 1234–1238. [Google Scholar]
  122. Liang, F.; Huo, W.; Ouyang, G.; Ji, P.; Wang, Z.; Wang, L.; Ye, X.-M. Effects of different types of exercise on motor function in patients with Chronic Kidney Disease. Chin. J. Rehabil. Theory Pract. 2018, 24, 208–213. [Google Scholar]
  123. Zhang, B.; Wu, C.; Gui, P.; Al, E. Effect of structured exercise training on old patients with Chronic Kidney Disease complicated with sarcopenia. Chin. J. Rehabil. Theory Pract. 2019, 25, 1463–1468. [Google Scholar]
  124. Li, J.X.; Hong, Y.; Chan, K.M. Tai chi: Physiological characteristics and beneficial effects on health. Br. J. Sports Med. 2001, 35, 148–156. [Google Scholar] [CrossRef] [PubMed]
  125. Shin, J.H.; Lee, Y.; Kim, S.G.; Choi, B.Y.; Lee, H.S.; Bang, S.Y. The beneficial effects of Tai Chi exercise on endothelial function and arterial stiffness in elderly women with rheumatoid arthritis. Arthritis Res. Ther. 2015, 17, 380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Janmaat, C.J.; van Diepen, M.; van Hagen, C.C.; Rotmans, J.I.; Dekker, F.W.; Dekkers, O.M. Decline of kidney function during the pre-dialysis period in Chronic Kidney Disease patients: A systematic review and meta-analysis. Clin. Epidemiol. 2018, 10, 613–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. O’Hare, A.M.; Batten, A.; Burrows, N.R.; Pavkov, M.E.; Taylor, L.; Gupta, I.; Todd-Stenberg, J.; Maynard, C.; Rodriguez, R.A.; Murtagh, F.E.; et al. Trajectories of Kidney Function Decline in the 2 Years Before Initiation of Long-term Dialysis. Am. J. Kidney Dis. 2012, 59, 513–522. [Google Scholar] [CrossRef] [Green Version]
  128. Lewis, E.J.; Hunsicker, L.G.; Bain, R.P.; Rohde, R.D. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N. Engl. J. Med. 1993, 329, 1456–1462. [Google Scholar] [CrossRef]
  129. Ruggenenti, P.; Perna, A.; Gherardi, G.; Garini, G.; Zoccali, C.; Salvadori, M.; Scolari, F.; Schena, F.P.; Remuzzi, G. Renoprotective properties of ACE-inhibition in non-diabetic nephropathies with non-nephrotic proteinuria. Lancet 1999, 354, 359–364. [Google Scholar] [CrossRef]
  130. Brenner, B.M.; Cooper, M.E.; de Zeeuw, D.; Keane, W.F.; Mitch, W.E.; Parving, H.H.; Remuzzi, G.; Snapinn, S.M.; Zhang, Z.; Shahinfar, S. Effects of Losartan on Renal and Cardiovascular Outcomes in Patients with Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2001, 345, 861–869. [Google Scholar] [CrossRef] [Green Version]
  131. Agodoa, L.Y.; Appel, L.; Bakris, G.L.; Beck, G.; Bourgoignie, J.; Briggs, J.P.; Charleston, J.; Cheek, D.; Cleveland, W.; Douglas, J.G.; et al. Effect of ramipril vs. amlodipine on renal outcomes in hypertensive nephrosclerosis: A randomized controlled trial. JAMA 2001, 285, 2719–2728. [Google Scholar] [CrossRef] [Green Version]
  132. Perkovic, V.; Jardine, M.J.; Neal, B.; Bompoint, S.; Heerspink, H.J.; Charytan, D.M.; Edwards, R.; Agarwal, R.; Bakris, G.; Bull, S.; et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2019, 380, 2295–2306. [Google Scholar] [CrossRef] [Green Version]
  133. Heerspink, H.J.L.; Stefánsson, B.V.; Correa-Rotter, R.; Chertow, G.M.; Greene, T.; Hou, F.F.; Mann, J.F.E.; McMurray, J.J.V.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef] [PubMed]
  134. Castaneda, C.; Gordon, P.L.; Parker, R.C.; Uhlin, K.L.; Roubenoff, R.; Levey, A.S. Resistance Training to Reduce the Malnutrition-Inflammation Complex Syndrome of Chronic Kidney Disease. Am. J. Kidney Dis. 2004, 43, 607–616. [Google Scholar]
  135. Gregory, S.M.; Headley, S.A.; Germain, M.; Flyvbjerg, A.; Frystyk, J.; Coughlin, M.A.; Milch, C.M.; Sullivan, S.; Nindl, B.C. Lack of circulating bioactive and immunoreactive IGF-I changes despite improved fitness in chronic kidney disease patients following 48 weeks of physical training. Growth Horm. IGF Res.: Off. J. Growth Horm. Res. Soc. Int. IGF Res. Soc. 2011, 21, 51–56. [Google Scholar]
  136. Headley, S.A.; Germain, M.; Wood, R.; Joubert, J.; Milch, C.; Evans, E.; Poindexter, A.; Cornelius, A.; Brewer, B.; Pescatello, L.S.; et al. Short-term aerobic exercise and vascular function in CKD stage 3: A randomized controlled trial. Am. J. Kidney Dis. 2014, 64, 222–229. [Google Scholar] [CrossRef] [Green Version]
  137. Headley, S.A.; Germain, M.; Wood, R.; Joubert, J.; Milch, C.; Evans, E.; Cornelius, A.; Brewer, B.; Taylor, B.; Pescatello, L.S. Blood pressure response to acute and chronic exercise in chronic kidney disease. Nephrology (Carlton, Vic.) 2017, 22, 72–78. [Google Scholar] [CrossRef]
  138. Zhou, Y.; Hellberg, M.; Hellmark, T.; Höglund, P.; Clyne, N. Twelve months of exercise training did not halt abdominal aortic calcification in patients with CKD - a sub-study of RENEXC-a randomized controlled trial. BMC Nephrol. 2020, 21, 233. [Google Scholar] [CrossRef]
  139. Small, D.M.; Beetham, K.S.; Howden, E.J.; Briskey, D.R.; Johnson, D.W.; Isbel, N.M.; Gobe, G.C.; Coombes, J.S. Effects of exercise and lifestyle intervention on oxidative stress in chronic kidney disease. Redox Rep.: Commun. Free Radic. Res. 2017, 22, 127–136. [Google Scholar] [CrossRef]
  140. Huppertz, N.; Beetham, K.S.; Howden, E.J.; Leicht, A.S.; Isbel, N.M.; Coombes, J.S. A 12-month lifestyle intervention does not improve cardiac autonomic function in patients with chronic kidney disease. Auton. Neurosci.: Basic Clin. 2020, 224, 102642. [Google Scholar] [CrossRef]
  141. Kirkman, D.L.; Ramick, M.G.; Muth, B.J.; Stock, J.M.; Pohlig, R.T.; Townsend, R.R.; Edwards, D.G. Effects of aerobic exercise on vascular function in nondialysis Chronic Kidney Disease: A randomized controlled trial. Am. J. Physiol. Ren. Physiol. 2019, 316, F898–F905. [Google Scholar] [CrossRef]
  142. Pechter, U.; Maaroos, J.; Mesikepp, S.; Veraksits, A.; Ots, M. Regular low-intensity aquatic exercise improves cardio-respiratory functional capacity and reduces proteinuria in chronic renal failure patients. Nephrol. Dial. Transplant. 2003, 18, 624–625. [Google Scholar] [CrossRef] [Green Version]
  143. Stevens, L.A.; Coresh, J.; Greene, T.; Levey, A.S. Assessing Kidney Function—Measured and Estimated Glomerular Filtration Rate. N. Engl. J. Med. 2006, 354, 2473–2483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Ferguson, T.W.; Komenda, P.; Tangri, N. Cystatin C as a biomarker for estimating glomerular filtration rate. Curr. Opin. Nephrol. Hypertens. 2015, 24, 295–300. [Google Scholar] [CrossRef] [PubMed]
  145. Stevens, L.A.; Schmid, C.H.; Greene, T.; Li, L.; Beck, G.J.; Joffe, M.M.; Froissart, M.; Kusek, J.W.; Zhang, Y.L.; Coresh, J.; et al. Factors other than glomerular filtration rate affect serum cystatin C levels. Kidney Int. 2009, 75, 652–660. [Google Scholar] [CrossRef] [Green Version]
  146. Chew-Harris, J.S.C.; Florkowski, C.M.; George, P.M.; Elmslie, J.L.; Endre, Z.H. The relative effects of fat versus muscle mass on cystatin C and estimates of renal function in healthy young men. Ann. Clin. Biochem. 2013, 50, 39–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Baxmann, A.C.; Ahmed, M.S.; Marques, N.C.; Menon, V.B.; Pereira, A.B.; Kirsztajn, G.M.; Heilberg, I.P. Influence of muscle mass and physical activity on serum and urinary creatinine and serum cystatin C. Clin. J. Am. Soc. Nephrol. CJASN 2008, 3, 348–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Macdonald, J.; Marcora, S.; Jibani, M.; Roberts, G.; Kumwenda, M.; Glover, R.; Barron, J.; Lemmey, A. GFR Estimation Using Cystatin C Is Not Independent of Body Composition GFR Estim. Using Cystatin C Is Not Indep. Body Compos. 2006, 48, 712–719. [Google Scholar]
  149. Kazeminia, M.; Daneshkhah, A.; Jalali, R.; Vaisi-Raygani, A.; Salari, N.; Mohammadi, M. The Effect of Exercise on the Older Adult’s Blood Pressure Suffering Hypertension: Systematic Review and Meta-Analysis on Clinical Trial Studies. Int. J. Hypertens. 2020, 2020, 2786120. [Google Scholar] [CrossRef]
  150. Naci, H.; Salcher-Konrad, M.; Dias, S.; Blum, M.R.; Sahoo, S.A.; Nunan, D.; Ioannidis, J.P.A. How Does Exercise Treatment Compare with Antihypertensive Medications? A Network Meta-Analysis of 391 Randomised Controlled Trials Assessing Exercise and Medication Effects on Systolic Blood Pressure. Br. J. Sport. Med. 2019, 53, 859–869. [Google Scholar] [CrossRef] [Green Version]
  151. Umpierre, D. Physical Activity Advice Only or Structured Exercise Training and Association with HbA 1c Levels in Type 2 Diabetes. JAMA 2011, 305, 1790. [Google Scholar] [CrossRef] [Green Version]
  152. Zhao, X.; He, Q.; Zeng, Y.; Cheng, L. Effectiveness of combined exercise in people with type 2 diabetes and concurrent overweight/obesity: A systematic review and meta-analysis. BMJ Open 2021, 11, e046252. [Google Scholar]
  153. Bird, S.R.; Hawley, J.A. Update on the effects of physical activity on insulin sensitivity in humans. BMJ Open Sport Exerc. Med. 2017, 2, e000143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Twells, L.K.; Harris Walsh, K.; Blackmore, A.; Adey, T.; Donnan, J.; Peddle, J.; Ryan, D.; Farrell, A.; Nguyen, H.; Gao, Z.; et al. Nonsurgical weight loss interventions: A systematic review of systematic reviews and meta-analyses. Obes. Rev. Off. J. Int. Assoc. Study Obes. 2021, 22, e13320. [Google Scholar] [CrossRef] [PubMed]
  155. Thompson, S.; Wiebe, N.; Padwal, R.S.; Gyenes, G.; Headley, S.A.; Radhakrishnan, J.; Graham, M. The effect of exercise on blood pressure in Chronic Kidney Disease: A systematic review and meta-analysis of randomized controlled trials. PLoS ONE 2019, 14, e0211032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Wu, L.; Liu, Y.; Wu, L.; Yang, J.; Jiang, T.; Li, M. Effects of exercise on markers of inflammation and indicators of nutrition in patients with Chronic Kidney Disease: A systematic review and meta-analysis. Int. Urol. Nephrol. 2021, 54, 815–826. [Google Scholar] [CrossRef]
  157. Van Huffel, L.; Tomson, C.R.; Ruige, J.; Nistor, I.; Van Biesen, W.; Bolignano, D. Dietary restriction and exercise for diabetic patients with Chronic Kidney Disease: A systematic review. PLoS ONE 2014, 9, e113667. [Google Scholar] [CrossRef] [Green Version]
  158. Sakamoto, K.; Goodyear, L.J. Invited review: Intracellular signaling in contracting skeletal muscle. J. Appl. Physiol. 2002, 93, 369–383. [Google Scholar] [CrossRef]
  159. Hellsten, Y.; Nyberg, M. Cardiovascular adaptations to exercise training. Compr. Physiol. 2016, 6, 1–32. [Google Scholar]
  160. Rivilis, I.; Milkiewicz, M.; Boyd, P.; Goldstein, J.; Brown, M.D.; Egginton, S.; Hansen, F.M.; Hudlicka, O.; Haas, T.L. Differential involvement of MMP-2 and VEGF during muscle stretch- versus shear stress-induced angiogenesis. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H1430–H1438. [Google Scholar] [CrossRef] [Green Version]
  161. Brown, M.D.; Hudlicka, O. Modulation of physiological angiogenesis in skeletal muscle by mechanical forces: Involvement of VEGF and metalloproteinases. Angiogenesis 2003, 6, 1–14. [Google Scholar] [CrossRef]
  162. Linke, A.; Schoene, N.; Gielen, S.; Hofer, J.; Erbs, S.; Schuler, G.; Hambrecht, R. Endothelial dysfunction in patients with Chronic Heart Failure: Systemic effects of lower-limb exercise training. J. Am. Coll. Cardiol. 2001, 37, 392–397. [Google Scholar] [CrossRef] [Green Version]
  163. Høier, B.; Rufener, N.; Bojsen-Møller, J.; Bangsbo, J.; Hellsten, Y. The effect of passive movement training on angiogenic factors and capillary growth in human skeletal muscle. J. Physiol. 2010, 588, 3833–3845. [Google Scholar] [CrossRef] [PubMed]
  164. Hood, D.A. Mechanisms of exercise-induced mitochondrial biogenesis in skeletal muscle. Appl. Physiol. Nutr. Metab. Physiol. Appl. Nutr. Metab. 2009, 34, 465–472. [Google Scholar] [CrossRef] [PubMed]
  165. Tang, L.X.; Wang, B.; Wu, Z.K. Aerobic Exercise Training Alleviates Renal Injury by Interfering with Mitochondrial Function in Type-1 Diabetic Mice. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2018, 24, 9081–9089. [Google Scholar] [CrossRef] [PubMed]
  166. Gu, Q.; Zhao, L.; Ma, Y.P.; Liu, J.D. Contribution of mitochondrial function to exercise-induced attenuation of renal dysfunction in spontaneously hypertensive rats. Mol. Cell. Biochem. 2015, 406, 217–225. [Google Scholar] [CrossRef]
  167. Jorge, L.; Reinecke, N.; Da Silva, W.; Jorge, M.L.S.; Schor, N. Impact of different exercise training on mitochondrial in diabetic nephropathy. J. Diabetes Metab. Disord. Control 2021, 8, 79–85. [Google Scholar] [CrossRef]
  168. Trøseid, M.; Lappegård, K.T.; Claudi, T.; Damås, J.K.; Mørkrid, L.; Brendberg, R.; Mollnes, T.E. Exercise reduces plasma levels of the chemokines MCP-1 and IL-8 in subjects with the metabolic syndrome. Eur. Heart J. 2004, 25, 349–355. [Google Scholar] [CrossRef] [Green Version]
  169. Balan, E.; Diman, A.; Everard, A.; Nielens, H.; Decottignies, A.; Deldicque, L. Endurance training alleviates MCP-1 and TERRA accumulation at old age in human skeletal muscle. Exp. Gerontol. 2021, 153, 111510. [Google Scholar] [CrossRef]
  170. Baturcam, E.; Abubaker, J.; Tiss, A.; Abu-Farha, M.; Khadir, A.; Al-Ghimlas, F.; Al-Khairi, I.; Cherian, P.; Elkum, N.; Hammad, M.; et al. Physical exercise reduces the expression of RANTES and its CCR5 receptor in the adipose tissue of obese humans. Mediat. Inflamm. 2014, 2014, 627150. [Google Scholar] [CrossRef]
  171. Soltani, N.; Esmaeil, N.; Marandi, S.M.; Hovsepian, V.; Momen, T.; Shahsanai, A.; Kelishadi, R. Assessment of the Effect of Short-Term Combined High-Intensity Interval Training on TLR4, NF-κB and IRF3 Expression in Young Overweight and Obese Girls. Public Health Genom. 2020, 23, 26–36. [Google Scholar] [CrossRef]
  172. Petersen, A.M.W.; Pedersen, B.K. The anti-inflammatory effect of exercise. J. Appl. Physiol. 2005, 98, 1154–1162. [Google Scholar] [CrossRef] [Green Version]
  173. de Sousa, C.V.; Sales, M.M.; Rosa, T.S.; Lewis, J.E.; de Andrade, R.V.; Simões, H.G. The Antioxidant Effect of Exercise: A Systematic Review and Meta-Analysis. Sports Med. 2017, 47, 277–293. [Google Scholar] [CrossRef] [PubMed]
  174. Benatti, F.B.; Pedersen, B.K. Exercise as an anti-inflammatory therapy for rheumatic diseases—Myokine regulation. Nat. Rev. Rheumatol. 2015, 11, 86–97. [Google Scholar] [CrossRef] [PubMed]
  175. Wen, M.S.; Wang, C.Y.; Lin, S.L.; Hung, K.C. Decrease in irisin in patients with Chronic Kidney Disease. PLoS ONE 2013, 8, e64025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Ebert, T.; Focke, D.; Petroff, D.; Wurst, U.; Richter, J.; Bachmann, A.; Lössner, U.; Kralisch, S.; Kratzsch, J.; Beige, J.; et al. Serum levels of the myokine irisin in relation to metabolic and renal function. Eur. J. Endocrinol. 2014, 170, 501–506. [Google Scholar] [CrossRef] [Green Version]
  177. Miyamoto-Mikami, E.; Sato, K.; Kurihara, T.; Hasegawa, N.; Fujie, S.; Fujita, S.; Sanada, K.; Hamaoka, T.; Tabata, I.; Iemitsu, M. Endurance Training-Induced Increase in Circulating Irisin Levels Is Associated with Reduction of Abdominal Visceral Fat in Middle-Aged and Older Adults. PLoS ONE 2015, 10, e0120354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Peng, H.; Wang, Q.; Lou, T.; Qin, J.; Jung, S.; Shetty, V.; Li, F.; Wang, Y.; Feng, X.H.; Mitch, W.E.; et al. Myokine mediated muscle-kidney crosstalk suppresses metabolic reprogramming and fibrosis in damaged kidneys. Nat. Commun. 2017, 8, 1493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Watson, E.L.; Baker, L.A.; Wilkinson, T.J.; Gould, D.W.; Graham-Brown, M.P.; Major, R.W.; Ashford, R.U.; Philp, A.; Smith, A.C. Reductions in skeletal muscle mitochondrial mass are not restored following exercise training in patients with Chronic Kidney Disease. FASEB J. 2020, 34, 1755–1767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Abreu, C.C.; Cardozo, L.F.M.F.; Stockler-Pinto, M.B.; Esgalhado, M.; Barboza, J.E.; Frauches, R.; Mafra, D. Does resistance exercise performed during dialysis modulate Nrf2 and NF-κB in patients with Chronic Kidney Disease? Life Sci. 2017, 188, 192–197. [Google Scholar] [CrossRef]
  181. White, D.W.; Raven, P.B. Autonomic neural control of heart rate during dynamic exercise: Revisited. J. Physiol. 2014, 592, 2491–2500. [Google Scholar] [CrossRef]
  182. Mueller, P.J. Exercise training and sympathetic nervous system activity: Evidence for physical activity dependent neural plasticity. Clin. Exp. Pharmacol. Physiol. 2007, 34, 377–384. [Google Scholar] [CrossRef]
  183. Meredith, I.T.; Friberg, P.; Jennings, G.L.; Dewar, E.M.; Fazio, V.A.; Lambert, G.W.; Esler, M.D. Exercise training lowers resting renal but not cardiac sympathetic activity in humans. Hypertension 1991, 18, 575–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Khan, M.S.; Bakris, G.L.; Shahid, I.; Weir, M.R.; Butler, J. Potential Role and Limitations of Estimated Glomerular Filtration Rate Slope Assessment in Cardiovascular Trials: A Review. JAMA Cardiol. 2022, 7, 549–555. [Google Scholar] [CrossRef] [PubMed]
  185. Grams, M.E.; Sang, Y.; Ballew, S.H.; Matsushita, K.; Astor, B.C.; Carrero, J.J.; Chang, A.R.; Inker, L.A.; Kenealy, T.; Kovesdy, C.P.; et al. Evaluating glomerular filtration rate slope as a surrogate end point for ESKD in clinical trials: An individual participant meta-analysis of observational data. J. Am. Soc. Nephrol. 2019, 30, 1746–1755. [Google Scholar] [CrossRef] [PubMed]
  186. Inker, L.A.; Heerspink, H.J.; Tighiouart, H.; Levey, A.S.; Coresh, J.; Gansevoort, R.T.; Simon, A.L.; Ying, J.; Beck, G.J.; Wanner, C.; et al. GFR slope as a surrogate end point for kidney disease progression in clinical trials: A meta-analysis of treatment effects of randomized controlled trials. J. Am. Soc. Nephrol. 2019, 30, 1735–1745. [Google Scholar] [CrossRef] [PubMed]
  187. Hoefield, R.A.; Kalra, P.A.; Baker, P.; Lane, B.; New, J.P.; O’Donoghue, D.J.; Foley, R.N.; Middleton, R.J. Factors Associated with Kidney Disease Progression and Mortality in a Referred CKD Population. Am. J. Kidney Dis. 2010, 56, 1072–1081. [Google Scholar] [CrossRef]
  188. Yang, W.; Xie, D.; Anderson, A.H.; Joffe, M.M.; Greene, T.; Teal, V.; Hsu, C.Y.; Fink, J.C.; He, J.; Lash, J.P.; et al. Association of Kidney Disease Outcomes with Risk Factors for CKD: Findings From the Chronic Renal Insufficiency Cohort (CRIC) Study. Am. J. Kidney Dis. 2014, 63, 236. [Google Scholar] [CrossRef] [Green Version]
  189. Inker, L.A.; Eneanya, N.D.; Coresh, J.; Tighiouart, H.; Wang, D.; Sang, Y.; Crews, D.C.; Doria, A.; Estrella, M.M.; Froissart, M.; et al. New Creatinine- and Cystatin C–Based Equations to Estimate GFR without Race. N. Engl. J. Med. 2021, 385, 1737–1749. [Google Scholar] [CrossRef]
  190. Kirkman, D.L.; Bohmke, N.; Carbone, S.; Garten, R.S.; Rodriguez-Miguelez, P.; Franco, R.L.; Kidd, J.M.; Abbate, A. Exercise intolerance in kidney diseases: Physiological contributors and therapeutic strategies. Am. J. Physiol.-Ren. Physiol. 2021, 320, F161–F173. [Google Scholar] [CrossRef]
  191. Clarke, A.L.; Young, H.M.L.; Hull, K.L.; Hudson, N.; Burton, J.O.; Smith, A.C. Motivations and barriers to exercise in Chronic Kidney Disease: A qualitative study. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc.-Eur. Ren. Assoc. 2015, 30, 1885–1892. [Google Scholar] [CrossRef] [Green Version]
  192. Baker, L.A.; March, D.S.; Wilkinson, T.J.; Billany, R.E.; Bishop, N.C.; Castle, E.M.; Chilcot, J.; Davies, M.D.; Graham-Brown, M.P.M.; Greenwood, S.A.; et al. Clinical practice guideline exercise and lifestyle in Chronic Kidney Disease. BMC Nephrol. 2022, 23, 201–211. [Google Scholar] [CrossRef]
Table 3. Measures of compliance in exercise training studies.
Table 3. Measures of compliance in exercise training studies.
Study ReferenceSession Attendance ComplianceExercise Intensity ComplianceExercise Time ComplianceEffect on VO2Peak (mL/kg/min) *
Aoike 2015, 2018, Baria 2014, Gomes 2017 [102,104,108,112]84–87%Not reported78%2.3–5.4
Barcellos 2018 [113]Not reported aNot reportedNot reportedNot measured c
Greenwood 2015 [10]79%Not reportedNot reported5.7
Headley 2012 [13]83%650 kcal/week (no target)Not reported1.4
Headley 2014, 2017, Miele 2017 [111,136,137]95–97%Not reported78%1.6
Hiraki 2017 [109]Not reportedTarget steps metNot reportedNot measured
Howden 2013,2015 [8,120]Supervised: 70%Not reported57%2.8
Ikizler 2018 [12]Not reported bNot reportedNot reportedNo significant difference
Kirkman 2019 [141]92%“Completed as prescribed”“Completed as prescribed”2.09
Kiuchi 2017 [110]Not reportedNot reportedNot reportedNot measured
Leehey 2016 [107]Not reportedNot reportedNot reportedNo significant difference
Leehey 2009 [100]Not reportedNot reportedNot reportedNo significant difference
Van Craenenbroeck 2015 [11]95%101% of HR targetNot reported5.82
Measures of compliance in exercise training studies included in meta-analyses of effects on kidney function. Intensity compliance refers to measures such as energy expenditure and heart rate (HR). Time refers to time per exercise session; a “70% reached in 63.8%”; b “85% completed at least 50%”; c Improved sit-to-stand 30 s; * Numeric results indicate statistically significant findings in the exercise training group.
Table 4. Effect of Exercise on Blood Pressure in ND-CKD: summary of meta-analyses results.
Table 4. Effect of Exercise on Blood Pressure in ND-CKD: summary of meta-analyses results.
Review ReferenceSBP AnalysisResult (mmHg), ET vs. UC
Zhang 2019 [118]Final follow-up−5.61 (−8.99, −2.23)
Thompson 2019 [155]Final follow-up−4.33 (−9.04, 0.38)
Villanego 2020 [17]Baseline to follow-up change−1.68 (−6.80, 3.44)
Yamamoto 2021 [115]Final follow-up−0.75 (−1.24, −0.26)
Van den Wyngaert 2018 [119]Baseline to follow-up change1.22 (−4.45, 6.90)
Effects of exercise training on systolic blood pressure (SBP) as found in meta-analyses of studies in patients with non-dialysis CKD (ND-CKD). Results are the mean difference (plus 95% CI) between exercise training (ET) and ususal care (UC) groups, either the SBP result at final follow-up, or the change from baseline to final follow-up.
Table 5. Effect of Exercise on body mass index in ND-CKD: summary of meta-analyses results.
Table 5. Effect of Exercise on body mass index in ND-CKD: summary of meta-analyses results.
Review ReferenceBMI AnalysisResult (kg/m 2), ET vs. UC
Zhang 2019 [118]Final follow-up−1.32 (−2.39, −0.25)
Villanego 2020 [17]Baseline to follow-up change−0.89 (−1.47, −0.31)
Wu 2021 [156]Final follow-up−0.77 (−1.31, 0.23)
Yamamoto 2021 [115]Final follow-up−0.19 (−0.38, 0.0)
Effects of exercise training on body mass index (BMI) as found in meta-analyses of studies in patients with non-dialysis CKD (ND-CKD). Results are the mean difference (plus 95% CI) between exercise training (ET) and ususal care (UC) groups, either the BMI result at final follow-up, or the change from baseline to final follow-up.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Davies, M.; Sandoo, A.; Macdonald, J. The Role of Exercise Training in Delaying Kidney Function Decline in Non-Dialysis-Dependent Chronic Kidney Disease. Kidney Dial. 2022, 2, 262-286. https://doi.org/10.3390/kidneydial2020026

AMA Style

Davies M, Sandoo A, Macdonald J. The Role of Exercise Training in Delaying Kidney Function Decline in Non-Dialysis-Dependent Chronic Kidney Disease. Kidney and Dialysis. 2022; 2(2):262-286. https://doi.org/10.3390/kidneydial2020026

Chicago/Turabian Style

Davies, Mark, Aamer Sandoo, and Jamie Macdonald. 2022. "The Role of Exercise Training in Delaying Kidney Function Decline in Non-Dialysis-Dependent Chronic Kidney Disease" Kidney and Dialysis 2, no. 2: 262-286. https://doi.org/10.3390/kidneydial2020026

Article Metrics

Back to TopTop