Exercise in The Prevention and Treatment of Type 2 Diabetes: Didactic Synopsis
Exercise in The Prevention and Treatment of Type 2 Diabetes: Didactic Synopsis
See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Exercise in the Prevention and Treatment of
Type 2 Diabetes
John P. Kirwan,* Elizabeth C. Heintz, Candida J. Rebello, and Christopher L. Axelrod
ABSTRACT
Type 2 diabetes is a systemic, multifactorial disease that is a leading cause of morbidity and
mortality globally. Despite a rise in the number of available medications and treatments avail-
able for management, exercise remains a first-line prevention and intervention strategy due to
established safety, efficacy, and tolerability in the general population. Herein we review the pre-
disposing risk factors for, prevention, pathophysiology, and treatment of type 2 diabetes. We
emphasize key cellular and molecular adaptive processes that provide insight into our evolving
understanding of how, when, and what types of exercise may improve glycemic control. © 2023
American Physiological Society. Compr Physiol 13:1-27, 2023.
we provide insight on how, when, and what types of exercise to TCF7L2, PPARG, IRS1, IGF1, CPT1A, and KCNQ1,
may improve glycemic control, compare exercise to gold among others (24, 185, 186). Furthermore, recent studies
standard and standard-of-care therapies, and address possible have investigated the role of ethnicity on type 2 diabetes
and known interactions between multiple treatment methods. incidence and heritability, specifically for minority popu-
lations in the United States and the United Kingdom, to
better characterize disease risk in historically understudied
Pathophysiology of Type 2 Diabetes populations (1, 65, 98).
training in patients with diabetes for 6 months, concluded Exercise timing may also have an impact on associated
that combination exercise conferred greater benefits and disease improvements. A randomized cross-over trial found
induced significantly more robust decreases in HbA1c than that HIIT in the afternoon had greater impact on lowering
either exercise type alone (269). Likewise, a follow-up study, blood glucose than morning HIIT sessions (257). Another
the Health Benefits of Aerobic and Resistance Exercise study found that postdinner exercise improved postprandial
Training in individuals with type 2 diabetes (HART-D) trail glucose and triglycerides, while predinner exercise only
controlled for work intensity between groups, and discovered improved glucose levels, indicating that postfeeding exercise
that aerobic exercise is more efficacious than resistance may be more optimal for improving diabetes symptoms
exercise in terms of HbA1c reduction, but the combination (119). While more research is needed on how timing impacts
of both aerobic and resistance exercise was still more ben- the benefits of exercise, especially with larger-scale studies,
eficial (52). Interestingly, the Resistance versus Aerobic exercise timing should be a consideration when planning and
Exercise training in type 2 diabetes (RAED2) trial showed evaluating the impacts of exercise interventions, as it may
that both resistance and aerobic exercise training for 4 months influence outcomes.
induced similar increases in hyperinsulinemic-euglycemic
clamp-derived insulin sensitivity while functional improve-
ments such as VO2max and maximum strength were specific Exercise as a prevention approach for type 2
to each training group (15). However, as VO2max is an diabetes
independent and significant predictor of insulin sensitivity In general, regular exercise improves cardiorespiratory fit-
(278), aerobic exercise is often assumed to be slightly more ness, lipid profiles, and glycemic control in individuals in
beneficial than resistance exercise in regulating diabetes both healthy and diseased states (183). A meta-analysis
symptoms. Nevertheless, the impacts of resistance exercise evaluating physical activity and diabetes incidence found that
cannot be discounted, as resistance exercise still confers there is a nonlinear relationship between exercise and disease
benefits in glucose and insulin signaling and is necessary for outcomes, with the greatest benefits of exercise occurring in
the maintenance of muscle mass and hypertrophy. Though those who were previously sedentary (273). Similarly, another
both aerobic and resistance exercise alone are capable of study on exercise and disease incidence reported that the rela-
lowering HbA1c , greatest improvements seem to occur with tionship between exercise frequency and lower disease risk
combination training (52, 269). Thus, the American Diabetes follows a J-shaped curve, with even moderate levels of phys-
Association (ADA) recommends combined aerobic and resis- ical activity significantly reducing diabetes risk in healthy
tance exercise for controlling glucose and lipids in diabetes individuals (155). Specifically, 16 weeks of exercise training
(58). However, more research is needed on the additive and improved insulin sensitivity in a dose-dependent manner
synergistic effects of combination exercise training (159). independent of age and gender, with no evidence of a maxi-
Collectively, despite disparities in the types and intensities mal dose response to exercise (70), providing strong evidence
of exercise that are best for treating and preventing diabetes, that exercise conveys health benefits and may prevent
some form of regular physical activity is far more favorable disease development even in healthy individuals. Further-
to none. more, endurance exercise training improved adipose tissue
It is also important to consider exercise intensity when insulin sensitivity and metabolic flexibility in healthy men
examining health outcomes associated with exercise, as and is associated with better insulin profiles and responses
intensity has been shown to greatly impact exercise benefits, compared to untrained healthy individuals (78, 245).
despite exercise modality. For example, aerobic high-intensity While exercise training appears to be an effective preven-
interval training (HIIT) has been reported to be more effec- tion strategy for healthy individuals, there is also evidence
tive than continuous aerobic training in both db/db mice that exercise may slow or even prevent diabetes onset in
and humans, with greater improvements in HbA1c , VO2max , patients who are overweight or have obesity and prediabetes.
body weight, and adipose tissue and skeletal muscle insulin In patients who are overweight, short-term exercise training
signaling resulting from interval training (45, 64, 302). Addi- has been shown to improve body mass index (BMI), fat mass,
tionally, interval training was more effective than continuous glucose tolerance, and insulin sensitivity, as well as reduce
training in improving lipoprotein profiles and reducing liver hepatic and skeletal muscle insulin resistance and C-reactive
lipid content in both prediabetic and patients with diabetes protein levels (115, 151, 193, 218). Additionally, VO2max has
despite similar alterations in substrate oxidation and lipid been shown to be inversely associated with HbA1c , fasting
metabolism (144, 211). Furthermore, interval training has glucose, and glucose tolerance responses in a mixed popula-
been shown to improve glucose effectiveness significantly tion of healthy individuals and patients with prediabetes or
more than continuous training in patients with diabetes diabetes (278), further suggesting that exercise may protect
(143); therefore, it is important to consider the intensity of against diabetes outcomes. Likewise, a meta-analysis on
exercise interventions when examining the corresponding adults with prediabetes found that long-term lifestyle inter-
impacts on metabolism and overall health improvements, as vention including regular exercise lowered diabetes incidence
higher intensity exercise appears to be associated with more in a 23-year follow-up (181), suggesting preventative actions
favorable outcomes. of exercise on diabetes development. Another meta-analysis
reported that 100 min/week of physical activity decreased phosphatidylinositol 3-kinase (PI3K) docking to IRS. PI3K
both fasting glucose and HbA1c in participants across the glu- stimulates membrane recruitment of phosphatidylinositol-
cose tolerance spectrum, with the positive effects of exercise 3,4,5-triphosphate (PIP3), which reciprocally recruits
being more profound in individuals with worse glucose phosphatidylinositol-3,4,5-triphosphate-dependent kinase
tolerance (29). Furthermore, in obese adults with prediabetes, (PDK1) and protein kinase B (AKT). PDK1 triggers a
12 weeks of exercise intervention enhanced β-cell function phosphorylation cascade, initiated with AKT and followed
and glucose-stimulated insulin secretion in association with by Akt substrate of 160 kDa (AS160), facilitating plasma
improvements in VO2max despite no change in body fat (194), membrane GLUT4 vesicle translocation, uptake of glucose,
providing evidence that exercise may play a significant role and glycogen synthesis (162, 244, 252). Preceding the onset
in mediating diabetes onset independent of weight loss. of diabetes, muscle cells become resistant to endogenous
Importantly, the beneficial effects of exercise to lower HbA1c insulin action while receptor and binding activity remain
occur in a dose-dependent manner, with 150 min/week of normal. This condition, known as insulin resistance, is an
physical activity inducing significantly greater improvements early pathogenic trait that contributes to hyperglycemia by
than exercising for less than 150 min/week (310). While this increasing the amount of insulin (and subsequently β-cell
highlights the added benefits of increased exercise duration, output) required to maintain plasma glucose homeostasis.
increasing the intensity of shorter exercise sessions has also Proximal signaling factors such as IRS-1, PI3K, AKT, and
been shown to have a similar effect on improving markers GLUT4 have augmented activity in patients with diabetes
of glycemic control in patients with prediabetes (64). Taken (132). These augmentations may entirely account for the
together, these findings highlight exercise for the prevention diminished sensitivity to insulin observed in patients with
and treatment of diabetes in both healthy and prediabetic IGT, prediabetes, or diabetes, though the net contribution
populations. remains somewhat unclear.
Exercise is one of the most rapid and effective means of
improving skeletal muscle insulin sensitivity (243), with
Impacts of Exercise on Glycemic Control measurable adaptation within days of initiating training (49).
Unlike endogenous insulin, which requires receptor signaling
in Type 2 Diabetes to activate AKT, muscle contraction can directly increase
Both aerobic and resistance exercise training have been glucose uptake via AMP-activated protein kinase (AMPK)
shown to improve glycemic control (15, 159, 269), and even activation of the GLUT4 cascade (26, 161, 221, 235). Like
small changes in habitual physical activity, such as increases any treatment, the effects of a single “dose” of exercise on
in daily walking by 400 steps, convey benefits on glucose insulin sensitivity dissipate within 48 to 72 h; thus, regular
regulation in patients with diabetes (332, 333). Exercise has and repeated exercise is necessary to induce chronic benefits
been shown to increase insulin action (117, 172), mediate in insulin sensitivity (40, 113, 117, 158, 322). Furthermore,
the secretion of cytokines such as interleukin 6 (IL-6) from the magnitude and extent of exercise-induced insulin sen-
skeletal muscle that can stimulate insulin secretion via GLP1 sitivity depend largely on exercise volume, intensity, and
production (77), improve β-cell function by reducing non- mode. While aerobic and resistance exercise increase insulin
pulsatile basal insulin secretion while enhancing pulsatile signaling via phosphorylation of AS160 and activation of
secretion (78), and resynchronize the circadian rhythmicity IRS-1 in skeletal muscle, the greatest effects are observed
of hormones and related physiology (328). Moreover, it with combination training (92, 130, 140).
can be broadly concluded that the benefits of exercise for
patients with diabetes can be achieved by aerobic, resistance,
and combination training (140), though different exercise
Aerobic exercise and glycemic control in type 2
modalities may have greater impacts on discrete metabolic diabetes
outcomes. Aerobic exercise facilitates numerous metabolic improve-
ments, including enhanced glucose regulation and insulin
sensitivity (Table 1), primarily through the recruitment of
Exercise training and insulin sensitivity in patients large muscle groups and associated cardiorespiratory func-
with type 2 diabetes tional improvements (120). Short-term increases in aerobic
Skeletal muscle glucose uptake is predominantly mediated by exercise via interval walking have been shown to improve
action of the insulin receptor, a concept that has been reviewed glucose effectiveness by approximately 50% in patients
extensively elsewhere (290). Rapid substrate exchange at with diabetes (143). However, while this robust improve-
the cellular level is maintained by a high affinity and sen- ment was associated with an amelioration of mean and
sitivity to the peptide hormone insulin and/or molecules peak glucose levels over a 24-h period, insulin sensitivity
mimicking its structure and activity. Insulin, insulin mimet- remained unchanged. Interestingly, longer intervention peri-
ics, and/or analogs activate the insulin receptor tyrosine ods report improvements in insulin sensitivity as well (145).
kinase, resulting in insulin receptor substrate (IRS) recruit- For example, 12 weeks of aerobic training has been shown to
ment and phosphorylation at multiple residues, triggering concurrently improve insulin sensitivity and reduce fasting
Aerobic exercise study Exercise prescription Duration Impact on glycemic control Secondary outcomes
Bacchi et al. (15) 60 min/day, 3 days/wk on cardiovascular training 4 months ↓ HbA1c , ↓ FPG, ↑ GDR ↑ VO2peak , ↓ BMI, ↓ fat mass, ↓ VAT, ↓ SAT,
equipment, progression to 60%–65% HRR ↑ lean mass
Church et al. (52) 12 kcal/kg at 50%–80% VO2max per wk 9 months ↔ HbA1c ↓ Waist circumference
Earnest et al. (73) 12 kcal/kg at 50%–80% VO2max per wk 9 months ↓ Metabolic syndrome score ↓ Waist circumference, ↓ SBP, ↑ VO2peak
Exercise and Type 2 Diabetes
Haus et al. (114) 1 h/day, 5 days/wk at 65% VO2max 12 wks ↓ Basal HGP, ↓ insulin-stimulated HGP, ↓ Body weight, ↓ abdominal fat, ↓ VAT
↓ hepatic insulin resistance
Jorge et al. (140) 1 h/day, 3 days/wk of cycling at HR corresponding 12 wks ↓ FPG, ↓ PPG, ↔ HbA1c , ↔ HOMA-IR, ↔ BMI, ↓ total cholesterol, ↓ TG, ↑ VO2peak ,
to lactate threshold ↑ IRS-1 ↓ blood pressure, ↓ c-reactive protein,
Karstoft et al. (144) 1 h/day, 5 days/wk of continuous (75%–80% 2 wks ↑ Glucose effectiveness, ↔ FPG, ↔ FPI ↔ Body mass, ↔ fat mass, ↔ RER, ↔ c-peptide
VO2max ) or interval (alternating 3 min sets of ∼60%
and ∼90% VO2max ) treadmill walking
Kasumov et al. (145) 1 h/day, 5 days/wk of treadmill or cycle ergometer, 12 wks ↓ Fasting insulin, ↑ glucose-stimulated ↓ Body weight, ↓ BMI, ↓ fat mass, ↓ TG, ↓ total
progression from 60% MHR to 80% MHR insulin disposal cholesterol, ↑ VO2max , ↓ plasma ceramides
Kirwan et al. (160) 50–60 min/day of cycling or treadmill walking at 7 days ↓ FPG, ↓ FPI, ↑ GDR, ↓ HGP ↔ Body weight, ↔ BMI, ↔ % body fat
80%–85% MHR
Ku et al. (168) 1 h/day, 5 days/wk of walking at 3–5 METs 12 wks ↔ HbA1c , ↔ FPG ↓ Body weight, ↓ fat mass, ↑ adiponectin,
↓ leptin, ↓ c-peptide
Li et al. (182) 30 min/day of cycle ergometer at 50%–70% MHR, 12 wks ↓ HbA1c , ↓ FPG, ↓ FPI ↑ VO2max (both groups), ↓ BMI, ↓ body mass
5 days/wk or 15 min/day (7 rounds of 1 min (continuous training only)
intervals at 80%–95% MHR followed by 1 min of
20%–30% MHR) of cycle ergometer, 5 days/wk
LookAHEAD Research 175 min/wk of moderate-intensity exercise 1 year ↓ HbA1c ↓ Body weight, ↓ waist circumference, ↔ CVD
Group (298) risk
Savikj et al. (257) ∼30 min/day (7 min warm-up followed by 6 rounds 2 wks ↔ HbA1c ↔ Total cholesterol, ↔ TG
of 1 min pulses over 220 W at 75 rpm with 1 min
rest between), 3 days/wk of cycling either in the
morning or afternoon
Sigal et al. (269) Treadmill or cycle ergometer, progression from 15 22 wks ↓ HbA1c ↓ Body weight, ↓ waist circumference, ↑ thigh
to 20 min/day at 60% MHR to 45 min/day at 75% muscle CSA, ↔ total cholesterol, ↔ blood
MHR, 3 days/wk pressure
Solomon et al. (276) 1 h/day, 5 days/wk at 60%–65% MHR; intensity 3 months ↓ FPG, ↓ 2 h OGTT, ↑ GDR, ↓ glucose ↓ Body weight, ↓ BMI, ↓ % body fat, ↓ VAT,
increased to 80%–85% at wk 4 AUC, ↑ insulin AUC, ↑ β-cell function ↓ leptin, ↓ TG, ↓ total cholesterol
Solomon et al. (277) 1 h/day, 4–5 days/wk of cycling or treadmill 12–16 wks ↔ HbA1c , ↓ FPG, ↓ 2 h OGTT, ↓ insulin ↓ Body weight, ↓ BMI, ↓ % body fat, ↑ VO2max
walking at ∼75% VO2max AUC
AUC, area under the curve; BMI, body mass index; FPG, fasting plasma glucose; FPI, fasting plasma insulin; GDR, glucose disposal rate; HbA1c , hemoglobin A1c ; HGP, hepatic glucose production;
HOMA-IR, homeostatic model assessment of insulin resistance; MHR, maximum heart rate; OGTT, oral glucose tolerance test; PPG, postprandial glucose; RER, respiratory exchange ratio; SAT,
subcutaneous adipose tissue; SBP, systolic blood pressure; TG, triglyceride; VAT, visceral adipose tissue.
Comprehensive Physiology
glucose, oral glucose tolerance, and HbA1c in patients with extent than aerobic or combined exercise training (Table 2).
diabetes (277). Furthermore, the benefits of resistance exercise are largely
While exercise is known to enhance GLP1 secretion in related to its role in preserving or increasing muscle mass and
healthy and obese individuals (122, 198), the impacts of physical function (25, 48).
exercise on GLP1 in patients with diabetes remains unclear
(107). Both aerobic HIIT and moderate-intensity continuous
training improved GLP1 secretion in men with obesity (200), Combination training and glycemic control in type 2
and short-term aerobic exercise training has been shown diabetes
to improve GLP1 sensitivity in patients with nonalcoholic Evidence supports aerobic and resistance training programs
fatty liver disease (NAFLD) (169), collectively indicating a for the greatest benefit in ameliorating diabetes symptoms,
favorable role of exercise in regulating GLP1. While acute as effects of both training modalities combined are additive
aerobic exercise did not enhance GLP1 secretion in patients (Table 3). Long-term combination training decreases HbA1c
with diabetes (80), both aerobic HIIT and low-intensity in patients with diabetes significantly more than either aero-
continuous exercise interventions induced improvements in bic or resistance exercise alone, in conjunction with inducing
GLP1 after a period of 12 weeks, with the greatest improve- greater improvements in clinical markers of metabolic
ments occurring after HIIT (175). Given the favorable syndrome (52, 73, 269). Similarly, 6 weeks of short-term
evidence around GLP1 in healthy individuals and those with functional high-intensity training improved β-cell function
obesity, it is plausible that long-term exercise intervention and insulin sensitivity with concurrent decreases in fat mass
may also enhance GLP1 secretion in patients with diabetes and preservation of lean mass (85, 219), and more recently,
as well. However, further research is needed to fully eluci- short-term combination aerobic and resistance exercise has
date the impacts of regular exercise on GLP1 secretion and been shown to significantly decrease HbA1c , fasting glucose,
to understand possible differential mechanistic actions of and fasting insulin in patients with diabetes (294). Impor-
exercise in healthy individuals and patients with diabetes. tantly, improvements in HbA1c with short-term combination
In contrast, gastric inhibitory polypeptide (GIP) secretion exercise intervention are associated with better mental health
seems to decrease with chronic aerobic exercise training, and quality of life in patients with diabetes (305). Longer-
although this effect may be dependent on combined lifestyle term combination training has also been shown to reduce
interventions (149, 197). Nevertheless, evidence suggests prescribed medication doses (139). Interestingly, differences
overall improvements in incretin secretion in response to in resistance training volume and intensity have not been
exercise; however, GIP remains understudied compared to shown to further impact improvements in HbA1c associated
GLP-1 in the context of diabetes. This is another area that with combination training (74, 331), although it is likely that
is ripe for future research and the opportunity to explore increases in resistance training volume or intensity in com-
the interactions of exercise and GLP-1 with GIP so as to bination with aerobic exercise would have additive benefits
better understand the overall impacts of exercise on incretin for secondary outcomes. Therefore, combined aerobic and
hormone secretion in patients with diabetes. resistance exercise induces greater enhancement of glycemic
control than either modality alone and thus is most effective
for managing diabetes symptoms (Figure 1).
Impacts of resistance exercise on clinical markers of
type 2 diabetes
Resistance exercise favors the neuromuscular system over Impact of exercise-associated weight loss on
cardiovascular adaptation (251). As such, improvements in glycemic control
glucose control are more directly attributable to improved Improvements in primary diabetes symptoms with exercise
substrate uptake and utilization, as the change in energy often occur in conjunction with weight loss and associated
expenditure is typically insufficient to decrease body weight improvements in body composition (15, 145, 277). Thus, it
(82, 236). The effects of resistance exercise in patients has been suggested that enhancements in glycemic control
with diabetes have been studied to a lesser extent than both with exercise training are the result of weight loss rather than
aerobic and combined exercise training; however, resis- stimulatory actions of exercise itself (276). Though it is often
tance training has been shown to mediate improvements difficult to distinguish improvements in glycemic control
in glucose tolerance (111, 119). For example, 16 weeks of associated with exercise from exercise-induced weight loss,
resistance exercise three times per week significantly lowered there are data to support exercise-induced enhancement of
HbA1c and the amount of medication required for diabetes glucose regulation independent of weight loss. One week of
control in high-risk older adults (41). Similarly, 12 weeks walking or cycling for 30 min/day increased glucose disposal
of moderate-intensity resistance training improved fasting rates and peripheral insulin sensitivity while reducing HGP
glucose, glycemic control, and components of the metabolic without inducing weight loss (160). Additionally, 12 weeks of
syndrome independent of body weight in adults with dia- HIIT training in men with diabetes improved HbA1c without
betes (205). Therefore, resistance training appears to convey inducing significant decreases in BMI or body mass (182).
benefits for glycemic control in diabetes, although to a lesser Contrarily, 12 weeks of moderate-intensity aerobic exercise
Resistance exercise study Exercise prescription Duration Impact on glycemic control Secondary outcomes
Bacchi et al. (15) 9 exercises/day on weight machines or with free 4 months ↓ HbA1c , ↓ FPG, ↑ GDR ↓ BMI, ↓ fat mass, ↓ VAT, ↓ SAT, ↑ lean mass,
weights, 3 sets of 10 reps, progression from 30% to ↑ leg extension 1RM, ↑ chest press 1RM
50% 1RM to 70%–80% 1RM
Castaneda et al. (41) 5 exercises/session on weight machines, 3 sets of 8 16 wks ↓ HbA1c , ↓ dose of prescribed T2D ↑ Muscle glycogen, ↑ lean mass, ↓ SBP, ↓ trunk
Exercise and Type 2 Diabetes
reps, progression from 60% to 80% baseline 1RM medication fat mass
to 70%–80% mid-study 1RM, 3 days/wk
Chen et al. (48) 5 exercises/day with resistance bands or isometric 12 wks ↔ HbA1c ↑ Muscle strength, ↑ balance, ↑ physical function
holds, 5 sets of 10 reps, 3 days/wk
Church et al. (52) 3 days/wk, 2 sets of 4 upper body exercises, 3 sets 9 months ↔ HbA1c ↑ Lean mass, ↓ fat mass, ↓ waist circumference
of 3 leg exercises, 2 sets of 2 core exercises,
10–12 reps/set
Earnest et al. (73) 3 days/wk, 2 sets of 4 upper body exercises, 3 sets 9 months ↔ Metabolic syndrome score ↓ Waist circumference
of 3 leg exercises, 2 sets of 2 core exercises,
10–12 reps/set
Hangping et al. (111) 4 isometric exercises/day on weight machines at 6 months ↓ HbA1c , ↓ FPG ↓ Total cholesterol, ↓ LDL,
progressively increasing maximal contraction for
5 s, 5–10 min/day, 1 day/wk
Heden et al. (119) 3 sets of 10 reps at RM of 8 upper and lower body Single bout ↓ Glucose AUC, ↓ insulin AUC ↓ TG AUC
exercises
Jorge et al. (140) full-body, 7-exercise circuit 3 days/wk 12 wks ↓ FPG, ↓ PPG, ↔ HbA1c , ↔ HOMA-IR, ↔ BMI, ↓ total cholesterol, ↓ TG
↓ c-reactive protein, ↑ IRS-1
Ku et al. (168) 3 sets of 15–20 reps at 40%–50% maximal 12 wks ↔ HbA1c , ↔ FPG ↓ Body weight, ↓ fat mass, ↑ adiponectin,
capacity of 10 upper and lower body exercises ↑ muscle mass, ↓ RBP4
with resistance bands, 5 days/wk
Misra et al. (205) 6 exercises/day at RM, 2 sets of 10 reps, 12 wks ↑ Insulin sensitivity, ↓ HbA1c , ↓ blood ↓ Total cholesterol, ↓ TG, ↓ SAT, ↔ BMI, ↔ lean
3 days/wk glucose, ↔ c-reactive protein mass, ↔ total body fat
Sigal et al. (269) 7 exercises/day on weight machines, progression 22 wks ↓ HbA1c ↓ Body weight, ↓ waist circumference, ↑ thigh
to 2–3 sets of 7–9 reps, 3 days/wk muscle CSA, ↔ total cholesterol, ↔ blood
pressure
1RM, one repetition maximum; AUC, area under the curve; BMI, body mass index; CSA, cross-sectional area; FPG, fasting plasma glucose; GDR, glucose disposal rate; HbA1c , hemoglobin
A1c ; HGP, hepatic glucose production; HOMA-IR, homeostatic model assessment of insulin resistance; LDL, low-density lipoprotein; RBP4, retinol-binding protein 4; RM, repetition maximum; SAT,
subcutaneous adipose tissue; SBP, systolic blood pressure; TG, triglyceride; VAT, visceral adipose tissue.
Comprehensive Physiology
Combination exercise study Exercise prescription Duration Impact on glycemic control Secondary outcomes
Church et al. (52) 10 kcal/kg at 50%–80% VO2max per wk plus 9 months ↓ HbA1c ↑ VO2max , ↓ body weight, ↓ fat mass, ↓ waist
2 days/wk, 1 set of 4 upper body exercises, 3 leg circumference
exercises and 2 core exercises, 10–12 reps/set
Earnest et al. (73) 10 kcal/kg at 50%–80% VO2max per wk plus 9 months ↓ Metabolic syndrome score ↑ VO2peak , ↓ waist circumference
2 days/wk, 1 set of 4 upper body exercises, 3 leg
exercises and 2 core exercises, 10–12 reps/set
Egger et al. (74) 2 days/wk of 1 h/day of cycle ergometer at 70% HRR 8 wks ↓ HbA1c , ↓ FPG ↓ Body weight, ↓ BMI, ↓ SAT, ↓ SBP, ↓ DBP
and 50 min/day of 6 exercises of either hypertrophy
(2 sets of 10–12 reps at 70% 1RM) or endurance
(2 sets of 20–30 reps at 40% 1RM) resistance exercise
on weight machines
Fealy et al. (85) 3 days/wk of CrossFit training >85% MHR for 6 wks ↑ Insulin sensitivity ↓ Fat mass, ↓ DBP, ↓ TG, ↑ fat oxidation,
8–20 min ↑ adiponectin
Johansen et al. (139) 30–60 min/day of aerobic exercise 5–6 days/wk, with 12 months ↔ HbA1c , ↓ 2 h OGTT, ↓ Body mass, ↓ BMI, ↓ fat mass, ↑ lean mass,
2–3 sessions/wk combined with resistance exercise ↓ prescribed T2D medication use ↓ abdominal fat, ↑ VO2max
Jorge et al. (140) 3 days/wk of alternating 1 h/day of cycling at HR 12 wks ↓ FPG, ↓ PPG, ↔ HbA1c , ↔ BMI, ↓ total cholesterol, ↓ TG
corresponding to lactate threshold or full-body, ↔ HOMA-IR, ↓ c-reactive protein,
7-exercise circuit ↑ IRS-1
Nieuwoudt et al. (219) 3 days/wk of CrossFit training with at least one session 6 wks ↔ FPG, ↔ FPI, ↑ β-cell function ↔ Body weight, ↓ fat mass, ↑ VO2max
at >85% MHR for 10–20 min
Sigal et al. (269) 3 days/wk of treadmill or cycle ergometer, progression 22 wks ↓ HbA1c ↓ Body weight, ↓ waist circumference, ↑ thigh
from 15 to 20 min/day at 60% MHR to 45 min/day at muscle CSA, ↔ cholesterol, ↔ blood pressure
75% MHR plus 7 exercises/day on weight machines,
progression to 2–3 sets of 7–9 reps
Terauchi et al. (294) 3 days/wk of aerobic (30 min/session at increasing 12 wks ↓ HbA1c , ↓ FPG, ↓ FPI, ↓ HOMA-IR ↓ HDL
intensity from light to vigorous) and resistance (at
repetition maximum) exercise
Tomas-Carus et al. (305) 25 min/day of aerobic exercise at 60%–65% MHR 12 wks ↓ HbA1c ↑ Maximal strength, ↑ physical function,
and 15 min/day of resistance exercise of 2–4 sets of ↑ mental health
10 reps, 3 days/wk
Yang et al. (331) 5 days/wk of walking at 75% HRR and 2 days/wk of 6 months ↓ HbA1c , ↓ FPI ↓ BMI, ↓ body fat, ↑ VO2peak
10 resistance exercises of 2–3 sets of 7–15 reps
1RM, one repetition maximum; AUC, area under the curve; BMI, body mass index; DBP, diastolic blood pressure; FPG, fasting plasma glucose; FPI, fasting plasma insulin; GDR, glucose disposal
rate; HbA1c , hemoglobin A1c ; HDL, high-density lipoprotein; HOMA-IR, homeostatic model assessment of insulin resistance; IRS-1, insulin receptor substrate 1; MHR, maximum heart rate; OGTT,
oral glucose tolerance test; PPG, postprandial glucose; RM, repetition maximum; SAT, subcutaneous adipose tissue; SBP, systolic blood pressure; T2D, type 2 diabetes; TG, triglyceride; VAT,
visceral adipose tissue.
9
Exercise and Type 2 Diabetes
10.1002/cphy.c220009, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cphy.c220009 by University Of Louisville, Wiley Online Library on [27/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10.1002/cphy.c220009, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cphy.c220009 by University Of Louisville, Wiley Online Library on [27/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Exercise and Type 2 Diabetes Comprehensive Physiology
Combination exercise
Exercise and skeletal muscle mass and function in
patients with type 2 diabetes
Insulin secretion
Exercise and skeletal muscle atrophy
Insulin sensitivity
Aerobic exercise Maintenance of muscle mass is an intricate balance between
anabolism and catabolism (89). In healthy skeletal muscle,
Hepatic glucose production degradation of damaged or denatured proteins is necessary
Resistance exercise
Glucose effectiveness for maintaining homeostasis (173, 179); however, dysregula-
tion of this homeostatic function induced by the acceleration
Figure 1 Exercise improves glucose homeostasis. Exercise improves
of protein degradation or reduced protein synthesis results in
glucose effectiveness by increasing insulin secretion and sensitivity while muscle atrophy (57, 81). Molecular mechanisms of muscle
lowering hepatic glucose production. Aerobic, resistance, and combi- wasting include inappropriately adapted anabolic growth
nation exercise training improve glucose homeostasis in varying magni- hormone and insulin-like growth factor-1 (IGF-1), as well as
tudes based on the volume, intensity, duration, and mode, with greatest
benefits seen with combination training. dysregulation of catabolic proteins such as tumor necrosis
factor-alpha (TNFα) and myostatin, can shift protein balance
toward muscle degradation (81). In atrophic muscles, ubiq-
in women with diabetes led to a lower BMI without altering uitin ligase activation regulates protein degradation through
glucose regulation or insulin sensitivity (168). While these subsequent activation of muscle RING-finger protein-1
differences may be due to variations in study design, sex, (MuRF-1) and atrogin-1 (28, 254). Simultaneously, down-
and race, determining the role of weight loss and exercise in regulation of the PI3K/AKT pathway can activate forkhead
mediating improvements in diabetes remains elusive as the transcription factors (FoxO), specifically FoxO1, leading
two are typically accomplished simultaneously. to an increase in transcription of MuRF-1 and atrogin-1
There is also evidence that the positive health outcomes (28, 254).
from exercise are associated more with weight loss than Type 2 diabetes can trigger muscle atrophy via prolonged
fitness (299); thus, it can also be argued that weight loss activation of degradative pathways, leading to reduced mus-
mediates the greatest effects of exercise on diabetes symp- cle content, quality, and function (89, 231). Insulin resistance
toms. Furthermore, there is substantial evidence that exercise is a key contributor to muscle atrophy, as it activates the
alleviates primary symptoms whether weight-loss mediated PI3K/AKT pathway and mammalian target of rapamycin
or not. While determining the weight-loss-dependent and (mTOR), which also drives glucose metabolism (171). Thus,
independent effects of exercise on glycemic control should impairments in this pathway have been implicated in reduced
be further examined to enhance current understandings of protein synthesis (17, 28). Likewise, insulin and IGF-1 act
the mechanistic effects of exercise as a treatment modality through the PI3K/AKT pathway to suppress FoxO1 activa-
for diabetes, weight loss is generally beneficial and recom- tion and downstream MuRF-1 and atrogin-1 transcription to
mended for patients who are overweight (326). Therefore, attenuate muscle wasting (254, 286). Patients with diabetes
regardless of whether the impacts of exercise on glucose are also reported to have 60% above normal expression of
regulation and insulin sensitivity are weight-loss mediated, FoxO1 (231), collectively indicating heightened atrophic
regular exercise is an effective treatment for diabetes. signaling in diabetes.
Exercise counteracts muscle atrophy and promotes muscle
hypertrophy via increased protein synthesis (3). Exercise
Impacts of Exercise on Comorbidities in activates extracellular receptor kinase (ERK) and mTOR
Patients with Type 2 Diabetes to increase ribosomal biogenesis and subsequently increase
the capacity for protein synthesis (86). Resistance exercise
While regular exercise training is effective for maintaining specifically has been shown to increase p70S6K, conse-
and regulating glucose homeostasis in patients with diabetes, quently activating the AKT/mTOR signaling pathway to
the powerful effects of exercise extend beyond glycemic induce anabolic processes in skeletal muscle (6, 99). Never-
control. Exercise enhances mitochondrial function, increases theless, both aerobic and resistance exercise have been shown
skeletal muscle mass and function, augments cardiovascular to prevent muscle wasting in typically atrophic conditions
health, attenuates inflammatory signaling, alleviates diabetic such as spaceflight and bedrest (3, 6), and thus, it is not sur-
neuropathies, and helps prevent diabetes-associated neu- prising that exercise can elucidate similar effects in diseased
rodegenerative diseases. In addition, exercise exerts positive states known to induce atrophy (321).
Lipolysis
Insulin Insulin
Glucose
Insulin IRS-
1
PI3K PI3K
Glucose
uptake Signal
cascade
Akt
Akt GLUT4
TNFα
TNFα
IL-6 IL-1β
IFN-γ IL-1β
mTOR
Figure 2 Mechanistic actions of exercise on the treatment of multiorgan symptoms of type 2 diabetes. Exercise
induces tissue-specific effects that contribute to improvements of whole-body markers of type 2 diabetes. Primary
systemic improvements are achieved via activated pathways in skeletal muscle, adipose tissue, and the car-
diorespiratory system. Exercise-mediated enhancements in glucose regulation, insulin sensitivity, and substrate
utilization at the tissue level contribute to the reduction of inflammatory cytokine production and systemic inflam-
mation and improvements in whole-body outcomes. AKT, protein kinase B; GLUT4, glucose transporter type 4;
IFN-γ, interferon gamma; IL-1β, interleukin 1 beta; IL-6, interleukin 6; IRS-1, insulin receptor substrate 1; PI3K,
phosphatidylinositol 3-kinase; mTOR, mammalian target of rapamycin; TNF-α, tumor necrosis factor-alpha.
In patients with diabetes, 6 weeks of resistance training improvements in muscle quality mediated improvements in
have been shown to induce qualitative changes in skeletal metabolic control (34). However, metabolic improvements
muscle and increases in muscle fiber diameter (125). Impor- in conjunction with increased muscle mass may depend on
tantly, these improvements are accompanied by increases disease severity. Another study found that resistance training
in the expression of proteins involved in skeletal muscle improved body composition and muscle mass across the
insulin signaling and glucose uptake, namely GLUT4 (125). glucose tolerance spectrum, but improvements in glycemic
Similar effects were seen after 16 weeks of strength training control seen in healthy individuals or those with prediabetes
in older adults with diabetes, as increases in muscle mass and were not observed in patients with diagnosed disease (97).
Thus, while exercise improves muscle mass and function shown that exercise intervention increased mitochondrial
in patients with diabetes, the evidence demonstrating these content in patients across the glucose tolerance spectrum
effects in conjunction with improvements in glycemic control without significantly impacting mitochondrial respiratory
are inconsistent. function (292). It has also been proposed that mitochondrial
Nevertheless, maintenance of muscle mass and prevention localization and membrane plasticity may contribute to the
of muscle wasting are crucial for many disease outcomes. bioenergetic adaptations to exercise (83), which is primarily
Although it may not significantly regulate improvements in achieved by the activation of fission and fusion machinery
glycemic control, resistance training is important for muscle (13). Impairments in mitochondrial quality control have been
mass and function. Given the known effects of aerobic and linked to skeletal muscle insulin sensitivity across the glucose
resistance exercise on glucose regulation and skeletal mus- tolerance continuum, with most favorable mitonuclear protein
cle maintenance, combined aerobic and resistance training balances and mitochondrial protein folding found in active
in patients with diabetes appears to be the most favorable individuals (129). Furthermore, twelve weeks of exercise
approach for maintaining muscle quality while also inducing training in previously sedentary patients with obesity resulted
favorable outcomes in glycemic control. This is particularly in reduced mitochondrial fission independent of changes in
important for older patients who face compounding effects of mitochondrial content (13, 84), suggesting a role for exercise
sarcopenia from both age and disease (18). Being overweight in mediating mitochondrial-induced improvements in dia-
induces progressive imbalance of muscle protein toward betes symptoms. However, recent findings also reveal the
breakdown in older adults, leading to decreased muscle impacts of race on the relationship between mitochondrial
content and quality (238), and diabetes further accelerates dynamics and insulin sensitivity, thus highlighting the need
age-related sarcopenia (17). However, long-term resistance for further investigation on the role of mitochondrial plasticity
training in older patients with diabetes increased muscle and diabetes development in diverse populations (69).
mass and decreased adiposity with associated reductions
in systemic inflammation and C-reactive protein (201).
Furthermore, combined aerobic and resistance training in Exercise and adipose tissue function
older patients with obesity led to maintenance of muscle
mass despite weight loss (59). Thus, exercise training elicits Derangements in adipose tissue insulin responsiveness and
metabolic improvements in older adults while also mediating inflammatory signaling, particularly visceral and ectopic fat,
increased or sustained muscle mass during aging, making are another pathological feature of diabetes. Adipose tissue
it an effective intervention for diabetes and age-related insulin resistance, the inability of insulin to inhibit lipolysis
sarcopenia. in adipocytes, is increased nearly threefold above normal in
patients with diabetes (96). A progressive decline in β-cell
function is associated with incremental increases in free fatty
acids (FFAs) and fasting adipose tissue insulin resistance (96).
Exercise and skeletal muscle mitochondrial plasticity
Additionally, GLP1 receptors that stimulate lipolysis corre-
Early studies on patients with diabetes revealed a number late positively with insulin resistance in patients with severe
of mitochondrial abnormalities in skeletal muscle including obesity (313). Consequently, this causes a cycle of dysregu-
reduced content, size, density, respiratory function, and lation, as increased FFA levels further contribute to the loss
subtype distribution (148, 187, 246). It was proposed that of glucose effectiveness in poorly controlled diabetes (116).
these changes resulted in sustained bioenergetic impair- Obesity-related diabetes is also characterized by alterations
ments, ultimately contributing to impaired insulin action. in adipose tissue cytokine secretion, namely overexpression
Reductions in skeletal muscle mitochondrial content and of TNFα, which is further associated with the development of
size have been consistently observed in obese patients with insulin resistance (128, 234, 311).
diabetes (118, 246), largely due to deficiencies in mitochon- Regular exercise is associated with numerous improve-
drial fission proteins and mitonuclear protein imbalances ments in adipose tissue that contribute to attenuating markers
(129). However, the functional intactness of mitochondria of diabetes. Although exercise mediates improvements in
remains highly debated, with contrasting reports of both regulating lipolysis and FFA release from adipose tissue
defective and intact respiratory activity (32, 206, 233). In (247), primary improvements in adipose tissue with exercise
contrast, exercise results in numerous structural and func- are via alterations in cytokine production. Chronic exercise
tional adaptations in mitochondria. These adaptations are training decreases adipose tissue inflammation, primar-
best characterized in skeletal muscle, where exercise in ily through decreased TNFα production (146, 223, 282,
patients with and without diabetes increases mitochondrial 327). Most notably, exercise has been shown to increase
number, size, oxidative capacity, electron transfer, and DNA adiponectin release and decrease leptin production in patients
(203, 303, 304). While determining whether the changes in with diabetes (115, 151, 168). Thus, exercise is capable of
mitochondrial function are independent of the increase in suppressing unfavorable adipose cytokine production while
mitochondrial content following aerobic and resistance train- stimulating secretion of favorable cytokines, all of which
ing was originally debated (110, 123, 124), it was recently contribute to adipose-related metabolic improvements.
Exercise and cardiorespiratory fitness in type 2 levels, including IL-6, IL-1β, and TNFα, numerous studies
diabetes show that long-term adaptations to exercise indicate an
overall decrease in inflammatory signaling (87, 88, 226, 282,
Insulin can promote myocardial regeneration directly (329) or 318). Importantly, physical activity status is the best predictor
indirectly via attenuation of systemic TNFα production (93). of inflammatory biomarker levels, specifically TNFα and
In view of the fact that diabetes is associated with decreases IL-6, even compared to other factors such as age (202, 285).
in insulin secretion, it is often associated with cardiac and In patients with diabetes, exercise training has been shown to
endothelial dysfunction (60), and evidence suggests a direct induce decreases in systemic inflammation that correspond
association with diastolic dysfunction that leads to cardio- with improvements in glucose regulation (280). Rodent
vascular disease and diminished cardiorespiratory function studies further support long-term exercise training as an anti-
across the glucose tolerance spectrum (314). Since vascular inflammatory regulator, as indicated by decreased cytokine
endothelial function is highly dependent on ambient hyper- production in skeletal muscle, heart, and liver tissues after
glycemia, hyperinsulinemia, and insulin resistance (190), prolonged exercise intervention in animals with diet-induced
multiple exercise models have been proposed to promote vas- obesity (141). Altogether, these findings demonstrate the
cular health in patients with diabetes (174, 297). Short-term positive impacts of exercise on systemic inflammation, which
exercise training rescued high-fat diet-induced loss of insulin- further extends to improvements in glycemic control and
stimulated glucose uptake in the myocardium and normalized insulin signaling in diabetes.
associated chemokine and cytokine levels in the heart (141),
and consistent long-term aerobic exercise has been shown to
enhance endothelial function (302). More recently, 3 months Exercise and diabetic neuropathies
of combined aerobic and resistance training in patients with Small and large nerve fibers play a role in tactile, vibration,
diabetes improved flow-mediated endothelium-dependent pain, and proprioceptive sensitivity (31). Diabetes can lead to
vasodilation and reduced cardiovascular events for 2 years impaired sensory function in these nerves, as hyperglycemia
compared to sedentary controls (222). Furthermore, regular plays a key role in the activation of biochemical pathways
exercise is known to improve cardiorespiratory fitness, thus that, together with impaired nerve perfusion, contribute to
contributing to improved cardiovascular health, and these the development of diabetic neuropathies (237, 337). Diag-
improvements have been linked to improvements in diastolic nosable neuropathy occurs in nearly half of all patients with
function and prevention of heart failure in patients with diabetes (72), and nerve conduction studies demonstrate that
diabetes (105). neuropathy is present in 10% to 18% of patients at the time
of diagnosis, suggesting that peripheral nerve injury occurs
early in the disease and coincides with the progression of
Impacts of exercise on systemic inflammation in type
insulin resistance (56). Diabetic neuropathy is characterized
2 diabetes by infections, ulcerations, and tissue destruction associated
Inflammation, especially as a result of tissue damage, is with neurological abnormalities and various levels of periph-
commonly associated with diseased states (47). Tissue eral artery disease that cause progressive disability and can
inflammation may play a role in the pathogenesis of diabetes, have consequences as severe as amputation (10, 11).
particularly obesity-associated diabetes, which appears to Regulation of glycemic control is the primary strategy
be mediated by oxidative stress, lipotoxicity, ectopic lipid for the prevention and management of diabetic neuropathies
deposition, and macrophage activation, most commonly in (266); however, pharmacological treatment of neuropathies
the liver and adipose tissue (67, 225, 268, 311). Long-term remains a challenge, highlighting the need for other treatment
immune system activation impairs insulin secretion and can strategies (337). Rational therapies aimed at direct control of
lead to diabetes and other comorbidities, including CVD (66, glucose have failed in preventing and treating diabetic neu-
75). Elevated levels of inflammatory markers and immune ropathy in human trials (271). In contrast, aerobic exercise
cells have been reported in humans and rodent models with training produces salutary effects simultaneously in many of
diabetes (76, 279). Furthermore, the European Prospec- the pathways that lead to neuropathy and has been shown
tive Investigation into Cancer and Nutrition (EPIC) study to improve symptoms of neuropathy and promote regrowth
reported that inflammatory cytokine levels, most notably of cutaneous small-diameter fibers in both animal models
IL-6 and TNFα, were elevated in patients with diabetes and human trials (61, 90, 126, 271). Additionally, mice were
(279). Moreover, IL-6 levels were identified as a predictor of able to regain islet function and demyelination of axons was
diabetes incidence independent of age, sex, BMI, and HbA1c prevented with exercise intervention during early stages of
(279), while inhibition of these cytokines has been shown to neuropathy (152). In addition to improving overall nerve
relieve β-cell dysfunction in human trials (75). integrity, exercise has also been shown to have positive
Exercise has chronic cytokine-lowering effects, and regular impacts on specific types of neuropathy. Diabetic retinopathy
physical activity elicits anti-inflammatory effects that account is the leading cause of visual impairment and blindness (287).
for decreased risk of chronic diseases (170, 270). Although Recent evidence suggests that while sedentary behavior is
acute responses to exercise actually include elevated cytokine associated with increased risk of retinopathy in diabetes,
physical activity is effective for preventing retinopathy in autonomic dysfunction and heart rate variability measures
high-risk patients and with improving vision in patients with were worse in diabetes, and these measures were associated
mild impairments (242). Similar effects have been reported with elevated HbA1c and diabetes duration (264).
for other forms of neuropathy, including two of the most The metabolic demands of exercise require sufficient
common in diabetes, diabetic peripheral neuropathy (DPN) increases in cardiac contractility and vagal innervation of the
and cardiac autonomic neuropathy (CAN). ventricles (274). Increased exercise capacity is associated
with reduced resting heart rate and increased cardiac vagal
activity, responses that are indicative of involvement of the
Exercise and diabetic peripheral neuropathy
parasympathetic branch of the autonomic nervous system
The most common form of neuropathy in diabetes is DPN, and suggest that parasympathetic function plays a role in
which is a chronic, symmetrical, length-dependent senso- exercise performance (101). In healthy untrained individuals,
rimotor polyneuropathy where autonomic dysfunction and aerobic exercise training reduces heart rate and increases
pain develop over time (295). Peripheral artery disease and vagal activity (4). Studies investigating the effects of exercise
neuropathy frequently coexist, and consequently, over 50% on cardiac autonomic function in healthy individuals and
of diabetic foot ulcers are ischemic or neuro-ischemic (239). patients with heart failure or diabetes demonstrate that the
Peripheral artery disease and collagen cross-links contribute speed of heart rate recovery is accelerated with exercise
to limited joint mobility, muscular impairment, and foot training (23, 62, 195, 288); thus, vagal tone can be enhanced
deformities, which in turn lead to changes in mechani- with exercise. Furthermore, regular exercise offers a means
cal properties of the tissues that can cause poor balance of training cardiac vagal tone, which in turn increases
(36, 90). Sensory loss often begins at the foot and progresses exercise capacity, thus amplifying cardiac improvements.
proximally (9). These impairments propagate a cycle that Consistent exercise may also modulate cardiac vagal tone
progressively alters foot plantar pressure distribution and via the enhancement of angiotensin II suppression (37).
exacerbation of the foot condition (90). Patients with diabetes Evidence suggests that both aerobic and resistance exercise
often have deficits in their ability to maintain posture prior may improve cardiac autonomic function in diabetes (20);
to ulceration and diagnosis of DPN (258). Small fiber injury however, few studies have investigated the effects of exercise
occurs early in disease progression and leads to neuropathic on cardiac function in this patient population. A recent meta-
pain, while overt gait and foot ulcers are due to loss of large analysis reported that exercise improved cardiac autonomic
myelinated fibers that regulate touch and proprioceptive function in diabetes, but the strength of the evidence was
function, functional abnormalities of DPN that are late com- low due to the scarcity of existing studies (23), highlighting
plications and reflect irreversible axonal loss (301). Regular the need for additional research examining the impacts of
exercise addresses glycemic control and other pathologies exercise on cardiac function in patients with diabetes to
associated with DPN, including microvascular function, determine the most impactful treatment methods.
lipid oxidation, and oxidative stress (61, 163, 271). More However, it is important to emphasize that exercise is
importantly, evidence suggests that axonal regeneration can intolerable for some patients with neuropathy and may not be
occur as a result of exercise training (90), thus preventing and a suitable treatment due to reduced heart rate, blood pressure,
relieving DPN symptoms. and cardiac output in response to exercise that is associated
with autonomic dysfunction (237). Moreover, in diabetes,
CAN typically occurs in conjunction with DPN, and pain
Impacts of exercise on cardiac autonomic
associated with DPN may further limit exercise capability
neuropathy in type 2 diabetes
(94). These conditions should be carefully considered when
In diabetes, CAN is defined as impairment of autonomic prescribing or studying exercise treatment, and appropriate
control of the cardiovascular system resulting from damage precautions should be employed. Additionally, there appears
to the nerves and blood vessels innervating the heart, which to be a point of no return where recovery and regeneration
can lead to dysfunctional heart rate control and abnormal of axonal function are not possible, particularly damage to
vascular dynamics (264). Chronic hyperglycemia leads to large myelinated axons (103, 152), making early intervention
progressive autonomic neural dysfunction that first affects crucial for prevention or reversal of neuropathic development.
the vagus nerve, which largely mediates parasympathetic
activity (272). In diabetes, this is characterized by initial
increases in sympathetic tone with increased resting heart Exercise and diabetic nephropathy
rate, while sympathetic denervation soon follows (14). CAN Diabetic nephropathy is a common long-term complication
is a complication that increases risk of all-cause mortality and the most common cause of mortality in patients with
and myocardial ischemia, a risk that is further heightened type 2 diabetes, as well as a leading cause of end-stage renal
in diabetes (199, 315). The Treatment Options for Type 2 failure globally (46, 253). Chronic dysregulation of glucose
Diabetes in Adolescents and Youth (TODAY) study, the homeostasis activates the renin-angiotensin-aldosterone sys-
largest reported cohort of participants with youth-onset T2D, tem, renal hyperfiltration, and oxidative stress, leading to renal
found that compared to participants with obesity, cardiac vascular abnormalities, glomerular lesions, and impairments
in glomerular filtration, resulting in kidney failure when left diabetes can cause cerebrovascular complications and lead
untreated (46, 189). Primary treatment methods are targeted to blood-brain barrier breakdown, initiating neural cell death
at improving glucose control and blood pressure regula- and dysfunction (21).
tion through antidiabetic pharmacotherapies or angiotensin Because of its chronic anti-inflammatory capabilities
receptor blockers to alleviate microvascular stress associated and associations with improved endothelial function and
with nephropathy, although continued disease progression increased brain capillarization, regular exercise has been
requires hemodialysis treatment, and in extreme cases, kidney proposed as a method to prevent neurodegenerative disease
transplant (253). However, some first-line pharmacological development in patients with diabetes (27, 191, 289, 319).
interventions for type 2 diabetes, including metformin, are Exercise training is also hypothesized to induce neurogenesis
contraindicated as a treatment for diabetic nephropathy, as (312), making it an appealing approach for the prevention
they are not metabolized by the liver and are thus excreted of metabolically associated neuronal diseases (21). In mice
by the kidneys, increasing renal stress (240). Hence, alter- genetically predisposed to the development of Alzheimer’s
native medications or adjustments to treatment strategies are disease, treadmill exercise ameliorated declining cognitive
required for simultaneous management of type 2 diabetes and function by suppressing neuronal cell death (165). Similarly,
complications from nephropathy. in diabetic rats, treadmill exercise was shown to prevent
The association of frailty and sarcopenia with mortality diabetes-associated neurodegenerative disease development
in patients with chronic kidney disease has increased inter- (154), collectively suggesting therapeutic effects of exercise
est in exercise as a preventative and long-term treatment on neurodegenerative disease development and progression.
to aid in survivability of renal-associated complications in While the majority of the evidence for exercise as a preven-
patients with type 2 diabetes (43, 127, 136, 138). Intradialytic tion and treatment for neurodegenerative diseases is based
exercise has been shown to improve hemodialysis effective- on preclinical rodent models, there is growing evidence that
ness (35, 131, 267), and regular exercise training is associated exercise prevents neurodegeneration in aging adults with
with improved survival in patients receiving hemodialysis diabetes. Six months of aerobic training has been shown
(209, 281). Although randomized controlled trials on exercise to improve executive function and lower β-amyloid levels
training and renal outcomes in patients with type 2 diabetes in older glucose-intolerant adults, suggesting cognitive-
are lacking, studies in diabetic preclinical animal models enhancing effects of aerobic exercise in older metabolically
support exercise training as a mechanistic regulator of renal impaired adults (16). Large-scale, long-term clinical studies
health through attenuation of renal apoptotic cell death, are needed to fully describe how exercise interventions
reduction in glomerular lesions, and reduced albumin excre- contribute to enhanced cognitive function in patients with
tion (306, 320, 330), supporting the use of exercise training diabetes and specifically explore the links between cognition,
as a potential mediator of kidney health in type 2 diabetes body composition, metabolism, and inflammation (335).
and highlighting the need for further investigation of the Nevertheless, evidence suggests that exercise may medi-
impacts of exercise training on renal outcomes in diabetic ate prevention and delay progression of neurodegenerative
patients. diseases in older patients with diabetes.
Effects of exercise on the development and Changes in microbial regulation with exercise in
progression of neurodegenerative diseases in type 2 type 2 diabetes
diabetes Given the recent interest in delivering personalized medicine
Patients with diabetes are at greater risk for developing neu- and findings regarding the impact of nutrition on the function
rodegenerative diseases such as Alzheimer’s and Parkinson’s of the gut and subsequent metabolic implications (51), the
or dementia (208, 250). Importantly, impaired glucose reg- effects and interactions of exercise on the gut microbiome
ulation via insulin resistance is a pathological characteristic are of growing interest. Diabetes is associated with distinct
that has been linked to increased risk of cognitive impairment microbial dysbiosis characterized by decreased butyrate-
and dementia (293). Obesity leads to neuroinflammatory producing bacteria, as well as increased pathogens and
molecular changes, specifically in the hypothalamus, thus microbes of oxidative stress resistance, compared to individu-
impairing neuroendocrine and autonomic regulation of glu- als without diabetes (241). While still a largely underexplored
cose homeostasis and insulin secretion (109). Hypothalamic field, preliminary studies show that exercise mediates alter-
inflammation links aging and diabetes to neurodegenerative ations in gut microbiota composition and function. Exercise
diseases (22, 177), and causal relationships between diabetes has been shown to improve gut microbiota to a greater extent
and neurodegenerative diseases have been demonstrated in than caloric restriction in rats with diet-induced obesity,
rodent models, as the development of Parkinson’s disease can independent of weight loss (324). In rats with early-onset
be promoted by excessive energy intake, and brain-insulin obesity, exercise similarly counteracted high-fat diet-induced
signaling changes and mitochondrial dysfunction can lead gut microbial imbalances and mediated intestinal barrier
to Huntington’s disease (33, 249, 262). Furthermore, inflam- preservation (38). Furthermore, seven days of vigorous aer-
matory, oxidative, and metabolic changes associated with obic exercise normalized GLP1 expression and responses
in patients with obesity and NAFLD (169), suggesting that metabolites mediating tissue crosstalk, specifically skeletal
exercise positively modulates gut-hormone regulation and muscle-initiated crosstalk (50). In conjunction with the pre-
may have implications for patients with diabetes. While viously reported impacts of exercise training on metabolomic
exploring the gut microbiota in the context of disease is still outcomes, these findings expand our understanding. How-
an emerging area of research, the few studies investigating the ever, they underscore the paramount importance of future
impacts of exercise on improving gut health in patients with larger clinical and translational trials that could characterize
diabetes show favorable results for using exercise to treat molecular determinants of the impacts of exercise in diabetes
microbial dysbiosis. In patients with diabetes, exercise has and other metabolic diseases (216).
been shown to reduce intestinal inflammatory markers, leaky
gut, and endotoxemia, thus improving overall gut microbial
symbiosis (210, 230); however, additional research is needed Genomic impacts of exercise on type 2 diabetes
to better understand the effects and mechanisms whereby Although genomic findings typically indicate disease risk and
exercise induces improvements in gut health. susceptibility rather than the cause, multiple genome-wide
association studies have identified and implicated the role
of various loci associated with diabetes. The Meta-Analysis
Impacts of exercise on metabolic profiles of patients of Glucose and Insulin Related Traits Consortium (MAGIC)
with type 2 diabetes study on genome-wide associations of glucose, insulin,
The effect of exercise on metabolomics has implications for β-cell function, and HOMA-IR in patients with diabetes
preventing metabolic-related disease outcomes, as exercise indicates that genes regulating glycemic traits can identify
training has been shown to induce changes in metabolite diabetes risk, specifically loci that modestly elevate fasting
signatures related to mitochondrial remodeling and improved glucose (71). Likewise, there is evidence that the phenotypic
cardiometabolic fitness in exercise-trained patients at risk responses to regular exercise in diabetes are heritable, and
for metabolic diseases compared to sedentary controls (133). the heterogeneity in metabolic responses to exercise training
Aerobic exercise has also been shown to alter the metabolome is partly explained by individual genetic structure, such as
of rats, and this occurs in a tissue-specific manner, explicitly differences in peroxisome proliferator-activated receptor
by increasing oxidative metabolism, acyl-carnitine metabo- gamma (PPARγ) isoforms or skeletal muscle glycogen syn-
lites, and tricarboxylic acid (TCA) cycle intermediates in the thase genes (106). Combined transcriptomic and metabolomic
liver and skeletal muscle (283). Additionally, both serum- analysis revealed that diabetes was associated with upreg-
and tissue-specific metabolomics may provide a unique ulation of glucose degradation and differential amino acid
opportunity to determine sensitive metabolite signatures of metabolism after exercise in comparison to healthy controls.
exercise and potentially identify metabolic responders and Furthermore, acute responses to exercise in patients with
nonresponders. Results of the Trainability and Metabolomics diabetes have been shown to induce compensatory regula-
(TIMES) study, which investigated the impact of continuous tion of amino acid biosynthesis genes (112). Thus, specific
or HIIT aerobic exercise in healthy, previously sedentary, exercise-regulated gene expression in diabetes may induce
young individuals for 8 weeks, identified potential character- alterations in glucose and amino acid metabolism. This is a
istics of exercise responses (42). High metabolic responders growing area of research, and the role of SNPs, mi-RNAs,
to exercise showed significant metabolomic differences lncRNAs, and other variants in the context of exercise training
at baseline compared to lower responders, specifically and diabetes are under active investigation.
higher concentrations of 3-hydroxybutyrate, glycerol, acetyl-
carnitine, alanine, proline, and pyruvate and lower levels of
isobutyrate, glycolate, lysine, and phenylalanine, collectively Exercise as an Adjunct Therapy for Type
suggesting that baseline amino acid metabolites may be
indicative of exercise training response potential (42).
2 Diabetes
More recent investigations have focused on the effects of While exercise training remains an effective treatment strat-
exercise on metabolomics in patients with diabetes and indi- egy and is still used as a primary intervention for diabetes,
cate alterations in lipid and amino acid metabolism (19, 104, medications such as metformin have become more com-
291). Studies show that there are significant metabolite dif- monly prescribed as first-line therapy. Moreover, exercise
ferences between patients with diabetes and normoglycemic is now suggested as an adjuvant therapy along with other
individuals, specifically in skeletal muscle (19). Interestingly, lifestyle interventions, medications, or surgical interventions.
these changes may begin to develop in independent tissues Thus, understanding the role of other treatment methods
prior to clinical diagnosis (316). Exercise interventions and the interactions with exercise is paramount to most
altered the metabolic profiles of patients across the glucose safely and effectively treat the disease. With the growing
tolerance spectrum, particularly ceramides, acylcarnitines, number of pharmacotherapies available and the increasing
and lipid profiles, with larger improvements in individuals prevalence and availability of various surgical techniques for
with glucose intolerance (121, 291). Furthermore, exercise patients with obesity and diabetes (176, 255), investigating
training in patients with diabetes led to improvements in the impacts of combined therapeutic methods is necessary for
understanding the additive or deleterious effects of multiple patients with prediabetes, when compared to lifestyle inter-
treatments combined. ventions, exercise is more efficacious in reducing long-term
incidence (215, 296). Because both metformin and exercise
activate AMPK, the combined effects of the two treatment
Efficacy of combined lifestyle interventions for the methods are expected to be additive. Surprisingly, studies
treatment of type 2 diabetes investigating these treatments in combination have reported
Combined lifestyle interventions are commonly recom- contradictory results. Acutely, combined exercise training
mended for preventing diabetes, typically via the inclusion and metformin were shown to lower postprandial glucose
of exercise and dietary modifications (256). Long-term com- more effectively than either therapy alone (79), and exercise
bination of diet and exercise for 6 years conferred lasting with metformin has been shown to enhance GLP1 and GIP
metabolic benefits for up to 23 years in terms of preventing secretion (80). Contrarily, other studies report that exercise
diabetes incidence (181). Numerous studies have examined counteracted metformin-induced improvements in glycemic
the effects of various dietary interventions in combination responses to a mixed meal (30, 214), while metformin
with consistent exercise training, showing the importance reduced exercise-associated improvements in insulin sensi-
of combined lifestyle interventions for preventing disease tivity, VO2max , and skeletal muscle mitochondrial respiration
onset and maximizing improvements in disease symptoms. when combined with aerobic exercise training for 12 weeks
Unsurprisingly, exercise and caloric restriction have additive (164). Additionally, while 12 weeks of exercise training
impacts on inducing weight loss and improving insulin increased insulin sensitivity, metformin in combination with
resistance. Caloric restriction of approximately 500 kcal/day exercise blunted this effect (192). Aerobic training alone
combined with moderate-intensity exercise has been reported was the most effective for managing diabetes and preventing
to achieve greater weight loss and larger improvements in NAFLD in rats, while combining metformin and exercise
adipose insulin resistance and protection from lipid-induced negated improvements in exercise-restored complete mito-
hepatic insulin resistance than exercise alone (114). Addi- chondrial palmitate oxidation (184). Therefore, while both
tionally, exercise combined specifically with carbohydrate exercise and metformin can mediate prevention and treatment
restriction, induced greater improvements in glycemic control of diabetes, collectively there is not a significant additive
and endothelial function than just dietary intervention (91). benefit of combined metformin and exercise interventions.
There is also evidence that metabolic benefits of exercise can Future research should consider examination of the possible
be enhanced by altering the quality of carbohydrates rather mechanisms via which each may attenuate the effects of the
than solely restricting them, as diets with a low glycemic other as well as the impact of dosage and timing of each
index combined with aerobic exercise have been shown to treatment when combined.
have synergistic metabolic effects in terms of postprandial Liraglutide, a GLP1 analog, is another medication com-
hyperinsulinemia, incretin responses, and systemic anti- monly used for treating diabetes. It primarily lowers blood
inflammatory effects in obese patients with prediabetes glucose but has also been shown to lower blood pressure,
(150, 275). Collectively, dietary interventions combined with body weight, and incidence of adverse cardiovascular events
exercise training induce greater improvements for patients (196, 217). Aerobic and resistance exercise in combination
with diabetes than either lifestyle intervention alone, high- with liraglutide decreased HbA1c , fasting plasma glucose,
lighting the importance of both diet and exercise in metabolic blood pressure, and body weight in patients with obesity
function and symptom relief. and diabetes to a greater extent than either treatment alone
(204). Furthermore, combined therapies induced the greatest
improvements in insulin sensitivity and GLP1 production
Combination of exercise and pharmacotherapies for (204), indicating that liraglutide is a more favorable option
type 2 diabetes treatment when combined with exercise compared to metformin.
Recent pharmacological advances have led to new medica- Although additional pharmacotherapies exist, their interac-
tions for diabetes, making them the primary intervention for tions with exercise are largely unstudied and thus remain
treatment and even prevention. As the number of available unknown. Therefore, future research should investigate mech-
medications continues to grow, understanding their interac- anistic interactions between exercise therapy and medication
tions with commonly recommended lifestyle interventions, on glycemic control and secondary outcomes to determine
particularly exercise, is crucial to understand the potential of the best treatment combinations for diabetes.
medications to treat the disease and how to maximize their
effects for symptom relief.
Metformin is an established and widely used medication
Exercise after bariatric surgery in patients with
with proven effectiveness in diabetes prevention and treat- type 2 diabetes
ment (309). Metformin lowers blood glucose by decreasing Bariatric surgery is a highly effective treatment for patients
hepatic glucose production and increasing glucose disposal in with obesity and diabetes and has been shown to have substan-
skeletal muscle through activation of AMPK (134, 156, 213, tially greater improvements compared to intensive medical
336). While metformin is an effective preventive agent for therapy at 1-, 3-, and 5 years postsurgery (157, 259–261).
Bariatric surgery combined with medical therapy is also with other diseases. However, because diabetes is associated
more effective than either intervention alone at decreasing with a large range of comorbidities, future research should
and reversing hyperglycemia and at improving secondary focus on the appropriate exercise doses and intensities nec-
outcomes of diabetes (55, 260, 323), indicating favorable essary to safely implement exercise interventions and elicit
effects of surgery when combined with other treatment the greatest metabolic improvements in combination with
methods. Exercise has also been validated as a feasible and other health conditions. Nevertheless, exercise is feasible for
effective adjunct therapy for long-term health maintenance most pregnant women, elderly individuals, and patients with
in bariatric surgery patients (53). Moderate-intensity exercise additional comorbid diseases. Although each case should
after bariatric surgery facilitates continued improvements in be considered individually, especially in these populations,
postprandial glucose regulation and insulin responses while exercise can induce favorable health outcomes for patients
also inducing greater weight loss than surgery alone (265). with diabetes and special health considerations.
Furthermore, combined aerobic and resistance training after
bariatric surgery promotes sustained endothelial improve-
ments and decreased inflammatory markers postsurgery, as
well as mitigates losses in bone density induced by bariatric Conclusions and Future Research
surgery (63, 212). Additional benefits are seen in skeletal Exercise has been extensively studied as a therapeutic inter-
muscle mitochondria, with improvements in mitochondrial vention for diabetes and has been validated as a treatment
respiration and enzymatic activity independent of changes in not only for glycemic control but also for numerous diabetes-
mitochondrial density (54). Therefore, exercise is not only associated comorbidities. While the mechanisms through
feasible in patients after bariatric surgery, but it is benefi- which exercise elicits improvements in glucose uptake and
cial for the long-term maintenance of health improvements insulin effectiveness are well-studied, how exercise ame-
initiated by surgical intervention. liorates secondary negative health outcomes is less defined.
Thus, future studies should investigate the crosstalk between
organs during and postexercise to fully characterize the
Population-Specific Considerations for impacts of exercise on organ-specific and systemic metabolic
improvements. Additionally, attention should be focused on
Exercise Prescription in Patients with genetic variations in exercise responses in populations with
Type 2 Diabetes related chronic diseases, particularly pertaining to racial and
ethnic differences and associated exercise outcomes. Further-
While exercise is generally beneficial for patients with more, links between genetic variation, the gut microbiome,
diabetes, there are sub-populations that warrant special con- and exercise-associated alterations should be considered.
sideration, as exercise may increase risk of adverse health Finally, with the increasing prevalence of exercise prescrip-
events under certain circumstances. Additional attention tion as an adjuvant therapy, interactions between exercise
should be given when evaluating the safety, feasibility, and and the growing number of pharmaceutical and nutraceutical
effectiveness of exercise interventions in these populations, treatments should be evaluated to determine their effective-
specifically pregnant women, elderly patients, and individ- ness and safety for diabetes treatment and management.
uals with other comorbid diseases. Although studies on
exercise in pregnant women with diabetes are scarce, low-
to-moderate intensity exercise during pregnancy has been References
safely implemented and was shown to improve glycemic
1. Abate N, Chandalia M. The impact of ethnicity on type 2 diabetes.
control in women who developed gestational diabetes (12). J Diabetes Complicat 17: 39-58, 2003. DOI: 10.1016/S1056-
However, exercise needs to be further evaluated as a therapy 8727(02)00190-3.
2. Afroz-Hossain A, Dawkins M, Myers AK. Sleep and environmental
for pregnant women with preexisting diabetes to fully deter- factors affecting glycemic control in people with type 2 diabetes
mine its safety and efficacy as a treatment (232). In elderly mellitus. Curr Diab Rep 19: 40, 2019. DOI: 10.1007/s11892-019-
1159-9.
patients, monitored and properly scaled exercise interven- 3. Akima H, Hotta N, Sato K, Ishida K, Koike T, Katayama K. Cycle
tions are safe and effective for opposing the deleterious ergometer exercise to counteract muscle atrophy during unilateral lower
limb suspension. Aviat Space Environ Med 80: 652-656, 2009. DOI:
effects of sarcopenia on skeletal muscle and for improving 10.3357/ASEM.2399.2009.
glycemic control in patients with obesity and diabetes (39, 4. Al-Ani M, Munir SM, White M, Townend J, Coote JH. Changes in
R-R variability before and after endurance training measured by power
201). The feasibility of exercise interventions has also been spectral analysis and by the effect of isometric muscle contraction.
demonstrated in patients with chronic kidney disease and Eur J Appl Physiol 74: 397-403, 1996. DOI: 10.1007/BF02337719.
5. Aldiss P, Betts J, Sale C, Pope M, Budge H, Symonds ME. Exercise-
NAFLD and was shown to induce positive effects on both induced ‘browning’ of adipose tissues. Metabolism 81: 63-70, 2018.
muscle mass and glycemic control (169, 229, 321). Exercise DOI: 10.1016/j.metabol.2017.11.009.
6. Alkner BA, Tesch PA. Knee extensor and plantar flexor muscle size
has also been safely implemented in patients with diabetes and function following 90 days of bed rest with or without resistance
and hypertension, and induced metabolic and cardiovascular exercise. Eur J Appl Physiol 93: 294-305, 2004. DOI: 10.1007/s00421-
004-1172-8.
improvements (284). Collectively, these studies indicate the 7. American Diabetes Association. Standards of medical care in diabetes,
feasibility of exercise for patients with diabetes in conjunction 2019. Diabetes Care 42: S1-S193, 2019.
8. American Diabetes Association. Classification and diagnosis of dia- 29. Boniol M, Dragomir M, Autier P, Boyle P. Physical activity and
betes: Standards of medical care in diabetes—2021. Diabetes Care 44: change in fasting glucose and HbA1c: A quantitative meta-analysis
S15-S33, 2021. DOI: 10.2337/dc21-S002. of randomized trials. Acta Diabetol 54: 983-991, 2017. DOI:
9. Andersen H, Gadeberg PC, Brock B, Jakobsen J. Muscular 10.1007/s00592-017-1037-3.
atrophy in diabetic neuropathy: A stereological magnetic reso- 30. Boulé NG, Robert C, Bell GJ, Johnson ST, Bell RC, Lewanczuk
nance imaging study. Diabetologia 40: 1062-1069, 1997. DOI: RZ, Gabr RQ, Brocks DR. Metformin and exercise in type 2 dia-
10.1007/s001250050788. betes: Examining treatment modality interactions. Diabetes Care 34:
10. Apelqvist J. Diagnostics and treatment of the diabetic foot. Endocrine 1469-1474, 2011. DOI: 10.2337/dc10-2207.
41: 384-397, 2012. DOI: 10.1007/s12020-012-9619-x. 31. Boulton AJM, Vinik AI, Arezzo JC, Bril V, Feldman EL, Freeman R,
11. Apelqvist J, Bakker K, van Houtum WH, Schaper NC. Practical guide- Malik RA, Maser RE, Sosenko JM, Ziegler D. Diabetic neuropathies:
lines on the management and prevention of the diabetic foot. Diabetes A statement by the American Diabetes Association. Diabetes Care 28:
Metab Res Rev 24: S181-S187, 2008. DOI: 10.1002/dmrr.848. 956-962, 2005. DOI: 10.2337/diacare.28.4.956.
12. Artal R. Exercise: The alternative therapeutic intervention for gesta- 32. Boushel R, Gnaiger E, Schjerling P, Skovbro M, Kraunsøe R,
tional diabetes. Clin Obstet Gynecol 46: 479-487, 2003. Dela F. Patients with type 2 diabetes have normal mitochondrial
13. Axelrod CL, Fealy CE, Mulya A, Kirwan JP. Exercise training remod- function in skeletal muscle. Diabetologia 50: 790-796, 2007. DOI:
els human skeletal muscle mitochondrial fission and fusion machinery 10.1007/s00125-007-0594-3.
towards a pro-elongation phenotype. Acta Physiol (Oxf) 225: e13216, 33. Bousquet M, St-Amour I, Vandal M, Julien P, Cicchetti F,
2018. DOI: 10.1111/apha.13216. Calon F. High-fat diet exacerbates MPTP-induced dopaminergic
14. Axelrod S, Lishner M, Oz O, Bernheim J, Ravid M. Spectral analy- degeneration in mice. Neurobiol Dis 45: 529-538, 2012. DOI:
sis of fluctuations in heart rate: An objective evaluation of autonomic 10.1016/j.nbd.2011.09.009.
nervous control in chronic renal failure. Nephron 45: 202-206, 1987. 34. Brooks N, Layne JE, Gordon PL, Roubenoff R, Nelson ME, Castaneda-
DOI: 10.1159/000184117. Sceppa C. Strength training improves muscle quality and insulin sen-
15. Bacchi E, Negri C, Zanolin ME, Milanese C, Faccioli N, Trombetta M, sitivity in Hispanic older adults with type 2 diabetes. Int J Med Sci 4:
Zoppini G, Cevese A, Bonadonna RC, Schena F, Bonora E, Lanza M, 19-27, 2006.
Moghetti P. Metabolic effects of aerobic training and resistance train- 35. Brown PDS, Rowed K, Shearer J, MacRae JM, Parker K. Impact
ing in type 2 diabetic subjects. Diabetes Care 35: 676-682, 2012. DOI: of intradialytic exercise intensity on urea clearance in hemodial-
10.2337/dc11-1655. ysis patients. Appl Physiol Nutr Metab 43: 101-104, 2018. DOI:
16. Baker LD, Frank LL, Foster-Schubert K, Green PS, Wilkinson CW, 10.1139/apnm-2017-0460.
McTiernan A, Cholerton BA, Plymate SR, Fishel MA, Watson GS, 36. Brownlee M. The pathobiology of diabetic complications. Diabetes 54:
Duncan GE, Mehta PD, Craft S. Aerobic exercise improves cognition 1615-1625, 2005.
for older adults with glucose intolerance, a risk factor for Alzheimer’s 37. Buch AN, Coote JH, Townend JN. Mortality, cardiac vagal control and
disease. J Alzheimers Dis 22: 569-579, 2010. DOI: 10.3233/JAD-2010- physical training – What’s the link? Exp Physiol 87: 423-435, 2002.
100768. DOI: 10.1111/j.1469-445X.2002.tb00055.x.
17. Bassil MS, Gougeon R. Muscle protein anabolism in type 2 dia- 38. Carbajo-Pescador S, Porras D, García-Mediavilla MV, Martínez-Flórez
betes. Curr Opin Clin Nutr Metab Care 16: 83-88, 2013. DOI: S, Juarez-Fernández M, Cuevas MJ, Mauriz JL, González-Gallego
10.1097/MCO.0b013e32835a88ee. J, Nistal E, Sánchez-Campos S. Beneficial effects of exercise on
18. Bauer J, Morley JE, Schols AMWJ, Ferrucci L, Cruz-Jentoft AJ, gut microbiota functionality and barrier integrity, and gut-liver
Dent E, Baracos VE, Crawford JA, Doehner W, Heymsfield SB, Jatoi crosstalk in an in vivo model of early obesity and non-alcoholic
A, Kalantar-Zadeh K, Lainscak M, Landi F, Laviano A, Mancuso fatty liver disease. Dis Model Mech 12: dmm039206, 2019. DOI:
M, Muscaritoli M, Prado CM, Strasser F, von Haehling S, Coats 10.1242/dmm.039206.
AJS, Anker SD. Sarcopenia: A time for action. An SCWD Position 39. Cartee GD, Hepple RT, Bamman MM, Zierath JR. Exercise promotes
Paper. J Cachexia Sarcopenia Muscle 10: 956-961, 2019. DOI: healthy aging of skeletal muscle. Cell Metab 23: 1034-1047, 2016.
10.1002/jcsm.12483. DOI: 10.1016/j.cmet.2016.05.007.
19. Beckman JA, Hu J-R, Huang S, Farber-Eger E, Wells QS, Wang TJ, 40. Cartee GD, Young DA, Sleeper MD, Zierath J, Wallberg-Henriksson H,
Gerszten RE, Ferguson JF. Metabolomics reveals the impact of type Holloszy JO. Prolonged increase in insulin-stimulated glucose trans-
2 diabetes on local muscle and vascular responses to ischemic stress. port in muscle after exercise. Am J Physiol Endocrinol Metab 256:
Clin Sci 1979 (134): 2369-2379, 2020. DOI: 10.1042/CS20191227. E494-E499, 1989. DOI: 10.1152/ajpendo.1989.256.4.E494.
20. Bellavere F, Cacciatori V, Bacchi E, Gemma ML, Raimondo D, Negri 41. Castaneda C, Layne JE, Munoz-Orians L, Gordon PL, Walsmith J,
C, Thomaseth K, Muggeo M, Bonora E, Moghetti P. Effects of aerobic Foldvari M, Roubenoff R, Tucker KL, Nelson ME. A randomized
or resistance exercise training on cardiovascular autonomic function of controlled trial of resistance exercise training to improve glycemic
subjects with type 2 diabetes: A pilot study. Nutr Metab Cardiovasc control in older adults with type 2 diabetes. Diabetes Care 25:
Dis 28: 226-233, 2018. DOI: 10.1016/j.numecd.2017.12.008. 2335-2341, 2002. DOI: 10.2337/diacare.25.12.2335.
21. Bertram S, Brixius K, Brinkmann C. Exercise for the diabetic brain: 42. Castro A, Duft RG, Ferreira MLV, de Andrade ALL, Gáspari AF,
How physical training may help prevent dementia and Alzheimer’s de Marchi Silva L, de Oliveira-Nunes SG, Cavaglieri CR, Ghosh S,
disease in T2DM patients. Endocrine 53: 350-363, 2016. DOI: Bouchard C, Chacon-Mikahil MPT. Association of skeletal muscle
10.1007/s12020-016-0976-8. and serum metabolites with maximum power output gains in response
22. Beydoun MA, Beydoun H, Wang Y. Obesity and central obesity as risk to continuous endurance or high-intensity interval training programs:
factors for incident dementia and its sub-types: A systematic review The TIMES study – A randomized controlled trial. PLoS One 14:
and meta-analysis. Obes Rev Off J Int Assoc Study Obes 9: 204-218, e0212115, 2019. DOI: 10.1371/journal.pone.0212115.
2008. DOI: 10.1111/j.1467-789X.2008.00473.x. 43. Çeliker M, Selçuk MY, Olt S. Sarcopenia in diabetic nephropathy: a
23. Bhati P, Shenoy S, Hussain ME. Exercise training and cardiac cross-sectional study. Romanian J Intern Med Rev Roum Med Interne
autonomic function in type 2 diabetes mellitus: A systematic 56: 102-108, 2018. DOI: 10.2478/rjim-2018-0003.
review. Diabetes Metab Syndr Clin Res Rev 12: 69-78, 2018. DOI: 44. Center for Disease Control and Prevention. Diabetes Report
10.1016/j.dsx.2017.08.015. Card [Online]. 2021. https://www.cdc.gov/diabetes/library/reports/
24. Billings LK, Florez JC. The genetics of type 2 diabetes: what have reportcard.html [25 Aug. 2021].
we learned from GWAS? Ann N Y Acad Sci 1212: 59-77, 2010. DOI: 45. Chavanelle V, Boisseau N, Otero YF, Combaret L, Dardevet D,
10.1111/j.1749-6632.2010.05838.x. Montaurier C, Delcros G, Peltier SL, Sirvent P. Effects of high-
25. Binder EF, Yarasheski KE, Steger-May K, Sinacore DR, Brown intensity interval training and moderate-intensity continuous training
M, Schechtman KB, Holloszy JO. Effects of progressive resistance on glycaemic control and skeletal muscle mitochondrial function in
training on body composition in frail older adults: Results of a ran- db/db mice. Sci Rep 7: 204, 2017. DOI: 10.1038/s41598-017-00276-8.
domized controlled trial. J Gerontol Ser A 60: 1425-1431, 2005. DOI: 46. Chawla T, Sharma D, Singh A. Role of the renin angiotensin system
10.1093/gerona/60.11.1425. in diabetic nephropathy. World J Diabetes 1: 141-145, 2010. DOI:
26. Bird SR, Hawley JA. Update on the effects of physical activity on 10.4239/wjd.v1.i5.141.
insulin sensitivity in humans. BMJ Open Sport Exerc Med 2: e000143, 47. Chen GY, Nuñez G. Sterile inflammation: Sensing and reacting to dam-
2017. DOI: 10.1136/bmjsem-2016-000143. age. Nat Rev Immunol 10: 826-837, 2010. DOI: 10.1038/nri2873.
27. Boa BCS, Souza M das GC, Leite RD, da Silva SV, Barja-Fidalgo 48. Chen S-M, Shen F-C, Chen J-F, Chang W-D, Chang N-J. Effects of
TC, Kraemer-Aguiar LG, Bouskela E. Chronic aerobic exercise resistance exercise on glycated hemoglobin and functional performance
associated to dietary modification improve endothelial function and in older patients with comorbid diabetes mellitus and knee osteoarthri-
eNOS expression in high fat fed hamsters. PLoS One 9: e102554, tis: A randomized trial. Int J Environ Res Public Health 17: 224, 2020.
2014. DOI: 10.1371/journal.pone.0102554. DOI: 10.3390/ijerph17010224.
28. Bodine SC, Baehr LM. Skeletal muscle atrophy and the E3 ubiquitin 49. Chibalin AV, Yu M, Ryder JW, Song XM, Galuska D, Krook A,
ligases MuRF1 and MAFbx/atrogin-1. Am J Physiol Endocrinol Metab Wallberg-Henriksson H, Zierath JR. Exercise-induced changes
307: E469-E484, 2014. DOI: 10.1152/ajpendo.00204.2014. in expression and activity of proteins involved in insulin signal
transduction in skeletal muscle: Differential effects on insulin-receptor intracellular insulin- and exercise-recruitable transporter pools
substrates 1 and 2. Proc Natl Acad Sci U S A 97: 38-43, 2000. in skeletal muscle. J Biol Chem 265: 13427-13430, 1990. DOI:
50. Chorell E, Otten J, Stomby A, Ryberg M, Waling M, Hauksson J, 10.1016/S0021-9258(18)77362-6.
Svensson M, Olsson T. Improved peripheral and hepatic insulin sensi- 69. Dubé JJ, Collyer ML, Trant S, Toledo FGS, Goodpaster BH, Kershaw
tivity after lifestyle interventions in type 2 diabetes is associated with EE, DeLany JP. Decreased mitochondrial dynamics is associ-
specific metabolomic and lipidomic signatures in skeletal muscle and ated with insulin resistance, metabolic rate, and fitness in African
plasma. Metabolites 11: 834, 2021. DOI: 10.3390/metabo11120834. Americans. J Clin Endocrinol Metab 105: 1210-1220, 2020. DOI:
51. Christiansen CB, Gabe MBN, Svendsen B, Dragsted LO, Rosenkilde 10.1210/clinem/dgz272.
MM, Holst JJ. The impact of short-chain fatty acids on GLP-1 70. Dubé JJ, Fleishman K, Rousson V, Goodpaster BH, Amati F. Exercise
and PYY secretion from the isolated perfused rat colon. Am J dose and insulin sensitivity: Relevance for diabetes prevention.
Physiol Gastrointest Liver Physiol 315: G53-G65, 2018. DOI: Med Sci Sports Exerc 44: 793-799, 2012. DOI: 10.1249/MSS.
10.1152/ajpgi.00346.2017. 0b013e31823f679f.
52. Church TS, Blair SN, Cocreham S, Johannsen N, Johnson W, Kramer 71. Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson
K, Mikus CR, Myers V, Nauta M, Rodarte RQ, Sparks L, Thompson A, AU, Wheeler E, Glazer NL, Bouatia-Naji N, Gloyn AL, Lindgren
Earnest CP. Effects of aerobic and resistance training on hemoglobin CM, Mägi R, Morris AP, Randall J, Johnson T, Elliott P, Rybin D,
A1c levels in patients with type 2 diabetes. J Am Med Assoc 304: 2253- Thorleifsson G, Steinthorsdottir V, Henneman P, Grallert H, Dehghan
2262, 2010. DOI: 10.1001/jama.2010.1710. A, Hottenga JJ, Franklin CS, Navarro P, Song K, Goel A, Perry JRB,
53. Coen PM, Carnero EA, Goodpaster BH. Exercise and bariatric surgery: Egan JM, Lajunen T, Grarup N, Sparsø T, Doney A, Voight BF,
An effective therapeutic strategy. Exerc Sport Sci Rev 46: 262-270, Stringham HM, Li M, Kanoni S, Shrader P, Cavalcanti-Proença C,
2018. DOI: 10.1249/JES.0000000000000168. Kumari M, Qi L, Timpson NJ, Gieger C, Zabena C, Rocheleau G,
54. Coen PM, Menshikova EV, Distefano G, Zheng D, Tanner CJ, Standley Ingelsson E, An P, O’Connell J, Luan J, Elliott A, McCarroll SA,
RA, Helbling NL, Dubis GS, Ritov VB, Xie H, Desimone ME, Smith Payne F, Roccasecca RM, Pattou F, Sethupathy P, Ardlie K, Ariyurek
SR, Stefanovic-Racic M, Toledo FGS, Houmard JA, Goodpaster BH. Y, Balkau B, Barter P, Beilby JP, Ben-Shlomo Y, Benediktsson R,
Exercise and weight loss improve muscle mitochondrial respiration, Bennett AJ, Bergmann S, Bochud M, Boerwinkle E, Bonnefond A,
lipid partitioning, and insulin sensitivity after gastric bypass surgery. Bonnycastle LL, Borch-Johnsen K, Böttcher Y, Brunner E, Bumpstead
Diabetes 64: 3737-3750, 2015. DOI: 10.2337/db15-0809. SJ, Charpentier G, Chen YDI, Chines P, Clarke R, Coin LJM, Cooper
55. Coen PM, Tanner CJ, Helbling NL, Dubis GS, Hames KC, Xie H, MN, Cornelis M, Crawford G, Crisponi L, Day INM, de Geus E,
Eid GM, Stefanovic-Racic M, Toledo FGS, Jakicic JM, Houmard JA, Delplanque J, Dina C, Erdos MR, Fedson AC, Fischer-Rosinsky A,
Goodpaster BH. Clinical trial demonstrates exercise following bariatric Forouhi NG, Fox CS, Frants R, Franzosi MG, Galan P, Goodarzi MO,
surgery improves insulin sensitivity. J Clin Invest 125: 248-257, 2015. Graessler J, Groves CJ, Grundy S, Gwilliam R, Gyllensten U, Hadjadj
DOI: 10.1172/JCI78016. S, Hallmans G, Hammond N, Han X, Hartikainen A-L, Hassanali N,
56. Cohen JA, Jeffers BW, Faldut D, Marcoux M, Schrier RW. Risks Hayward C, Heath SC, Hercberg S, Herder C, Hicks AA, Hillman DR,
for sensorimotor peripheral neuropathy and autonomic neuropathy in Hingorani AD, Hofman A, Hui J, Hung J, Isomaa B, Johnson PRV,
non-insulin-dependent diabetes mellitus (NIDDM). Muscle Nerve 21: Jørgensen T, Jula A, Kaakinen M, Kaprio J, Kesaniemi YA, Kivimaki
72-80, 1998. DOI: 10.1002/(SICI)1097-4598(199801)21:1<72::AID- M, Knight B, Koskinen S, Kovacs P, Kyvik KO, Lathrop GM, Lawlor
MUS10>3.0.CO;2-2. DA, Le Bacquer O, Lecoeur C, Li Y, Lyssenko V, Mahley R, Mangino
57. Cohen S, Nathan JA, Goldberg AL. Muscle wasting in disease: Molec- M, Manning AK, Martínez-Larrad MT, McAteer JB, McCulloch LJ,
ular mechanisms and promising therapies. Nat Rev Drug Discov 14: McPherson R, Meisinger C, Melzer D, Meyre D, Mitchell BD, Morken
58-74, 2015. DOI: 10.1038/nrd4467. MA, Mukherjee S, Naitza S, Narisu N, Neville MJ, Oostra BA, Orrù
58. Colberg SR, Sigal RJ, Yardley JE, Riddell MC, Dunstan DW, Dempsey M, Pakyz R, Palmer CNA, Paolisso G, Pattaro C, Pearson D, Peden JF,
PC, Horton ES, Castorino K, Tate DF. Physical activity/exercise and Pedersen NL, Perola M, Pfeiffer AFH, Pichler I, Polasek O, Posthuma
diabetes: A position statement of the American Diabetes Association. D, Potter SC, Pouta A, Province MA, Psaty BM, Rathmann W, Rayner
Diabetes Care 39: 2065-2079, 2016. DOI: 10.2337/dc16-1728. NW, Rice K, Ripatti S, Rivadeneira F, Roden M, Rolandsson O,
59. Colleluori G, Aguirre L, Phadnis U, Fowler K, Armamento-Villareal R, Sandbaek A, Sandhu M, Sanna S, Sayer AA, Scheet P, Scott LJ,
Sun Z, Brunetti L, Park JH, Kaipparettu BA, Putluri N, Auetumrong- Seedorf U, Sharp SJ, Shields B, Sigurðsson G, Sijbrands EJG, Silveira
sawat V, Yarasheski K, Qualls C, Villareal DT. Aerobic plus resistance A, Simpson L, Singleton A, Smith NL, Sovio U, Swift A, Syddall H,
exercise in obese older adults improves muscle protein synthesis and Syvänen A-C, Tanaka T, Thorand B, Tichet J, Tönjes A, Tuomi T,
preserves myocellular quality despite calorie restriction. Cell Metab Uitterlinden AG, van Dijk KW, van Hoek M, Varma D, Visvikis-Siest
30 (2): 261-273.e6, 2019. S, Vitart V, Vogelzangs N, Waeber G, Wagner PJ, Walley A, Walters
60. Cook CB, Hentz JG, Miller WJ, Tsui C, Naylor DB, Ziemer DC, Waller GB, Ward KL, Watkins H, Weedon MN, Wild SH, Willemsen G,
LA. Relationship of diabetes with cardiovascular disease-related hospi- Witteman JCM, Yarnell JWG, Zeggini E, Zelenika D, Zethelius B,
talization rates, length of stay, and charges: Analysis by race/ethnicity, Zhai G, Zhao JH, Zillikens MC, Borecki IB, Loos RJF, Meneton P,
age, and sex. Ethn Dis 17: 714-720, 2007. Magnusson PKE, Nathan DM, Williams GH, Hattersley AT, Silander
61. Cooper MA, Kluding PM, Wright DE. Emerging relationships between K, Salomaa V, Smith GD, Bornstein SR, Schwarz P, Spranger J,
exercise, sensory nerves, and neuropathic pain. Front Neurosci 10: 372, Karpe F, Shuldiner AR, Cooper C, Dedoussis GV, Serrano-Ríos
2016. DOI: 10.3389/fnins.2016.00372. M, Morris AD, Lind L, Palmer LJ, Hu FB, Franks PW, Ebrahim S,
62. Daanen HAM, Lamberts RP, Kallen VL, Jin A, Meeteren NLUV. A Marmot M, Kao WHL, Pankow JS, Sampson MJ, Kuusisto J, Laakso
systematic review on heart-rate recovery to monitor changes in training M, Hansen T, Pedersen O, Pramstaller PP, Wichmann HE, Illig T,
status in athletes. Int J Sports Physiol Perform 7: 251-260, 2012. DOI: Rudan I, Wright AF, Stumvoll M, Campbell H, Wilson JF, Hamsten
10.1123/ijspp.7.3.251. A, Bergman RN, Buchanan TA, Collins FS, Mohlke KL, Tuomilehto
63. Dantas WS, Gil S, Murai IH, Costa-Hong V, Peçanha T, Merege-Filho J, Valle TT, Altshuler D, Rotter JI, Siscovick DS, Penninx BWJH,
CAA, de Sá-Pinto AL, de Cleva R, Santo MA, Pereira RMR, Kirwan JP, Boomsma D, Deloukas P, Spector TD, Frayling TM, Ferrucci L, Kong
Roschel H, Gualano B. Reversal of improved endothelial function after A, Thorsteinsdottir U, Stefansson K, van Duijn CM, Aulchenko YS,
bariatric surgery is mitigated by exercise training. J Am Coll Cardiol Cao A, Scuteri A, Schlessinger D, Uda M, Ruokonen A, Jarvelin M-R,
72: 2278-2279, 2018. DOI: 10.1016/j.jacc.2018.07.094. Waterworth DM, Vollenweider P, Peltonen L, Mooser V, Abecasis GR,
64. De Nardi AT, Tolves T, Lenzi TL, Signori LU, da Silva AMV. Wareham NJ, Sladek R, Froguel P, Watanabe RM, Meigs JB, Groop
High-intensity interval training versus continuous training on physi- L, Boehnke M, McCarthy MI, Florez JC, Barroso I. New genetic loci
ological and metabolic variables in prediabetes and type 2 diabetes: implicated in fasting glucose homeostasis and their impact on type 2
A meta-analysis. Diabetes Res Clin Pract 137: 149-159, 2018. DOI: diabetes risk. Nat Genet 42: 105-116, 2010. DOI: 10.1038/ng.520.
10.1016/j.diabres.2017.12.017. 72. Dyck PJ, Kratz KM, Karnes JL, Litchy WJ, Klein R, Pach JM, Wil-
65. Divers J, Mayer-Davis EJ, Lawrence JM, Isom S, Dabelea D, Dolan son DM, O’Brien PC, Melton LJ. The prevalence by staged severity of
L, Imperatore G, Marcovina S, Pettitt DJ, Pihoker C, Hamman RF, various types of diabetic neuropathy, retinopathy, and nephropathy in
Saydah S, Wagenknecht LE. Trends in incidence of type 1 and type a population-based cohort: The Rochester Diabetic Neuropathy Study.
2 diabetes among youths — Selected counties and indian reservations, Neurology 43: 817-817, 1993. DOI: 10.1212/WNL.43.4.817.
United States, 2002–2015. Morb Mortal Wkly Rep 69: 161-165, 2020. 73. Earnest CP, Johannsen NM, Swift DL, Gillison FB, Mikus CR,
DOI: 10.15585/mmwr.mm6906a3. Lucia A, Kramer K, Lavie CJ, Church TS. Aerobic and strength
66. Donath MY, Dinarello CA, Mandrup-Poulsen T. Targeting innate training in concomitant metabolic syndrome and type 2 diabetes.
immune mediators in type 1 and type 2 diabetes. Nat Rev Immunol 19: Med Sci Sports Exerc 46: 1293-1301, 2014. DOI: 10.1249/MSS.
734-746, 2019. DOI: 10.1038/s41577-019-0213-9. 0000000000000242.
67. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. 74. Egger A, Niederseer D, Diem G, Finkenzeller T, Ledl-Kurkowski E,
Nat Rev Immunol 11: 98-107, 2011. DOI: 10.1038/nri2925. Forstner R, Pirich C, Patsch W, Weitgasser R, Niebauer J. Different
68. Douen AG, Ramlal T, Rastogi S, Bilan PJ, Cartee GD, Vranic types of resistance training in type 2 diabetes mellitus: Effects on gly-
M, Holloszy JO, Klip A. Exercise induces recruitment of the caemic control, muscle mass and strength. Eur J Prev Cardiol 20: 1051-
“insulin-responsive glucose transporter”. Evidence for distinct 1060, 2013. DOI: 10.1177/2047487312450132.
75. Eguchi K, Manabe I. Macrophages and islet inflammation in health in adults with metabolic syndrome: A three-month random-
type 2 diabetes. Diabetes Obes Metab 15: 152-158, 2013. DOI: ized controlled trial. J Sci Med Sport 25: 579-585, 2022. DOI:
10.1111/dom.12168. 10.1016/j.jsams.2022.04.002.
76. Ehses JA, Perren A, Eppler E, Ribaux P, Pospisilik JA, Maor-Cahn 96. Gastaldelli A, Gaggini M, DeFronzo RA. Role of adipose tissue insulin
R, Gueripel X, Ellingsgaard H, Schneider MKJ, Biollaz G, Fontana resistance in the natural history of type 2 diabetes: Results from the
A, Reinecke M, Homo-Delarche F, Donath MY. Increased number of San Antonio metabolism study. Diabetes 66: 815-822, 2017. DOI:
islet-associated macrophages in type 2 diabetes. Diabetes 56: 2356- 10.2337/db16-1167.
2370, 2007. DOI: 10.2337/db06-1650. 97. Geirsdottir OG, Arnarson A, Briem K, Ramel A, Jonsson PV, Thors-
77. Ellingsgaard H, Hauselmann I, Schuler B, Habib AM, Baggio LL, dottir I. Effect of 12-week resistance exercise program on body com-
Meier DT, Eppler E, Bouzakri K, Wueest S, Muller YD, Hansen AMK, position, muscle strength, physical function, and glucose metabolism
Reinecke M, Konrad D, Gassmann M, Reimann F, Halban PA, Gro- in healthy, insulin-resistant, and diabetic elderly icelanders. J Gerontol
mada J, Drucker DJ, Gribble FM, Ehses JA, Donath MY. Interleukin-6 Ser A 67: 1259-1265, 2012. DOI: 10.1093/gerona/gls096.
enhances insulin secretion by increasing glucagon-like peptide-1 98. Goff LM. Ethnicity and type 2 diabetes in the UK. Diabet Med 38:
secretion from L cells and alpha cells. Nat Med 17: 1481-1489, 2011. 927-938, 2019.
DOI: 10.1038/nm.2513. 99. Gonzalez AM, Hoffman JR, Townsend JR, Jajtner AR, Wells AJ, Beyer
78. Engdahl JH, Veldhuis JD, Farrell PA. Altered pulsatile insulin secre- KS, Willoughby DS, Oliveira LP, Fukuda DH, Fragala MS, Stout JR.
tion associated with endurance training. J Appl Physiol 79: 1977-1985, Association between myosin heavy chain protein isoforms and intra-
1995. DOI: 10.1152/jappl.1995.79.6.1977. muscular anabolic signaling following resistance exercise in trained
79. Erickson ML, Little JP, Gay JL, McCully KK, Jenkins NT. Postmeal men. Physiol Rep 3: e12268, 2015. DOI: 10.14814/phy2.12268.
exercise blunts postprandial glucose excursions in people on metformin 100. Goodyear LJ, Kahn BB. Exercise, glucose transport, and insulin
monotherapy. J Appl Physiol 123: 444-450, 2017. DOI: 10.1152/jap- sensitivity. Annu Rev Med 49: 235-261, 1998. DOI: 10.1146/annurev.
plphysiol.00213.2017. med.49.1.235.
80. Eshghi SRT, Bell GJ, Boulé NG. Effects of aerobic exercise with or 101. Gourine AV, Ackland GL. Cardiac Vagus and Exercise. Physiology 34:
without metformin on plasma incretins in type 2 diabetes. Can J Dia- 71-80, 2019. DOI: 10.1152/physiol.00041.2018.
betes 37: 375-380, 2013. DOI: 10.1016/j.jcjd.2013.07.030. 102. Gregory JM, Slaughter JC, Duffus SH, Smith TJ, LeStourgeon LM,
81. Fanzani A, Conraads VM, Penna F, Martinet W. Molecular Jaser SS, McCoy AB, Luther JM, Giovannetti ER, Boeder S, Pettus
and cellular mechanisms of skeletal muscle atrophy: An update. JH, Moore DJ. COVID-19 severity is tripled in the diabetes community:
J Cachexia Sarcopenia Muscle 3: 163-179, 2012. DOI: 10.1007/s13539- A prospective analysis of the pandemic’s impact in type 1 and type 2
012-0074-6. diabetes. Diabetes Care 44: 526-532, 2021. DOI: 10.2337/dc20-2260.
82. Farinatti P, Castinheiras Neto AG, Amorim PR. Oxygen consumption 103. Griffin JW, Thompson WJ. Biology and pathology of nonmyelinating
and substrate utilization during and after resistance exercises performed Schwann cells. Glia 56: 1518-1531, 2008. DOI: 10.1002/glia.20778.
with different muscle mass. Int J Exerc Sci 9: 77-88, 2016. 104. Gu X, Al Dubayee M, Alshahrani A, Masood A, Benabdelka-
83. Fealy CE, Mulya A, Axelrod CL, Kirwan JP. Mitochondrial dynamics mel H, Zahra M, Li L, Abdel Rahman AM, Aljada A. Distinctive
in skeletal muscle insulin resistance and type 2 diabetes. Transl Res metabolomics patterns associated with insulin resistance and type
202: 69-82, 2018. DOI: 10.1016/j.trsl.2018.07.011. 2 diabetes mellitus [Online]. Front Mol Biosci 7: 609806, 2020.
84. Fealy CE, Mulya A, Kirwan JP. Lifestyle intervention reduces https://www.frontiersin.org/article/10.3389/fmolb.2020.609806 [19
skeletal muscle dynamin-related protein-1 (Drp1) activation in Jan. 2022].
obese insulin resistant humans. FASEB J 27: 1209.18, 2013. DOI: 105. Gulsin GS, Swarbrick DJ, Athithan L, Brady EM, Henson J, Baldry E,
10.1096/fasebj.27.1_supplement.1209.18. Argyridou S, Jaicim NB, Squire G, Walters Y, Marsh A-M, McAdam
85. Fealy CE, Nieuwoudt S, Foucher JA, Scelsi AR, Malin SK, Pagadala J, Parke KS, Biglands JD, Yates T, Khunti K, Davies MJ, McCann GP.
M, Cruz LA, Li M, Rocco M, Burguera B, Kirwan JP. Functional Effects of low-energy diet or exercise on cardiovascular function in
high-intensity exercise training ameliorates insulin resistance and working-age adults with type 2 diabetes: A prospective, randomized,
cardiometabolic risk factors in type 2 diabetes. Exp Physiol 103: open-label blinded end point trial. Diabetes Care 43: 1300-1310, 2020.
985-994, 2018. DOI: 10.1113/EP086844. DOI: 10.2337/dc20-0129.
86. Figueiredo VC, Caldow MK, Massie V, Markworth JF, Cameron- 106. Hagberg JM, Jenkins NT, Spangenburg E. Exercise training, genetics
Smith D, Blazevich AJ. Ribosome biogenesis adaptation in resistance and type 2 diabetes-related phenotypes. Acta Physiol 205: 456-471,
training-induced human skeletal muscle hypertrophy. Am J Physiol 2012. DOI: 10.1111/j.1748-1716.2012.02455.x.
Endocrinol Metab 309: E72-E83, 2015. DOI: 10.1152/ajpendo. 107. Hamasaki H. Exercise and glucagon-like peptide-1: Does exercise
00050.2015. potentiate the effect of treatment? World J Diabetes 9: 138-140, 2018.
87. Fischer CP. Interleukin-6 in acute exercise and training: What is the DOI: 10.4239/wjd.v9.i8.138.
biological relevance? Exerc Immunol Rev 12: 6-33, 2006. 108. Hamilton MT, Hamilton DG, Zderic TW. Sedentary behavior as a
88. Flynn MG, McFarlin BK, Phillips MD, Stewart LK, Timmerman KL. mediator of type 2 diabetes. Med Sport Sci 60: 11-26, 2014. DOI:
Toll-like receptor 4 and CD14 mRNA expression are lower in resistive 10.1159/000357332.
exercise-trained elderly women. J Appl Physiol 95: 1833-1842, 2003. 109. Han C, Rice MW, Cai D. Neuroinflammatory and autonomic mecha-
DOI: 10.1152/japplphysiol.00359.2003. nisms in diabetes and hypertension. Am J Physiol Endocrinol Metab
89. Franch HA, Price SR. Molecular signaling pathways regulating muscle 311: E32-E41, 2016. DOI: 10.1152/ajpendo.00012.2016.
proteolysis during atrophy. Curr Opin Clin Nutr Metab Care 8: 271- 110. Hancock CR, Han D-H, Chen M, Terada S, Yasuda T, Wright DC, Hol-
275, 2005. DOI: 10.1097/01.mco.0000165005.01331.45. loszy JO. High-fat diets cause insulin resistance despite an increase in
90. Francia P, Gulisano M, Anichini R, Seghieri G. Diabetic foot muscle mitochondria. Proc Natl Acad Sci 105: 7815-7820, 2008. DOI:
and exercise therapy: Step by step the role of rigid posture and 10.1073/pnas.0802057105.
biomechanics treatment. Curr Diabetes Rev 10: 86-99, 2014. DOI: 111. Hangping Z, Xiaona Q, Qi Z, Qingchun L, Na Y, Lijin J, Siying L,
10.2174/1573399810666140507112536. Shuo Z, Xiaoming Z, Xiaoxia L, Qian X, Jaimovich D, Yiming L,
91. Francois ME, Myette-Cote E, Bammert TD, Durrer C, Neudorf H, Bin L. The impact on glycemic control through progressive resistance
DeSouza CA, Little JP. Carbohydrate restriction with postmeal walk- training with bioDensityTM in Chinese elderly patients with type 2
ing effectively mitigates postprandial hyperglycemia and improves diabetes: The PReTTy2 (Progressive Resistance Training in Type 2
endothelial function in type 2 diabetes. Am J Physiol Heart Circ Diabetes) Trial. Diabetes Res Clin Pract 150: 64-71, 2019. DOI:
Physiol 314: H105-H113, 2018. DOI: 10.1152/ajpheart.00524.2017. 10.1016/j.diabres.2019.02.011.
92. Frøsig C, Rose AJ, Treebak JT, Kiens B, Richter EA, Wojtaszewski 112. Hansen JS, Zhao X, Irmler M, Liu X, Hoene M, Scheler M, Li Y,
JFP. Effects of endurance exercise training on insulin signaling in Beckers J, Hrabĕ de Angelis M, Häring H-U, Pedersen BK, Lehmann
human skeletal muscle: Interactions at the level of phosphatidylinositol R, Xu G, Plomgaard P, Weigert C. Type 2 diabetes alters metabolic
3-Kinase, Akt, and AS160. Diabetes 56: 2093-2102, 2007. DOI: and transcriptional signatures of glucose and amino acid metabolism
10.2337/db06-1698. during exercise and recovery. Diabetologia 58: 1845-1854, 2015. DOI:
93. Fu F, Zhao K, Li J, Xu J, Zhang Y, Liu C, Yang W, Gao C, Li J, Zhang 10.1007/s00125-015-3584-x.
H, Li Y, Cui Q, Wang H, Tao L, Wang J, Quon MJ, Gao F. Direct 113. Hansen PA, Nolte LA, Chen MM, Holloszy JO. Increased GLUT-4
evidence that myocardial insulin resistance following myocardial translocation mediates enhanced insulin sensitivity of muscle glucose
ischemia contributes to post-ischemic heart failure. Sci Rep 5: 17927, transport after exercise. J Appl Physiol 85: 1218-1222, 1998. DOI:
2015. DOI: 10.1038/srep17927. 10.1152/jappl.1998.85.4.1218.
94. Gandhi RA, Marques JLB, Selvarajah D, Emery CJ, Tesfaye S. Painful 114. Haus JM, Solomon TPJ, Marchetti CM, Edmison JM, González
diabetic neuropathy is associated with greater autonomic dysfunction F, Kirwan JP. Free fatty acid-induced hepatic insulin resistance is
than painless diabetic neuropathy. Diabetes Care 33: 1585-1590, 2010. attenuated following lifestyle intervention in obese individuals with
DOI: 10.2337/dc09-2314. impaired glucose tolerance. J Clin Endocrinol Metab 95: 323-327,
95. Garthwaite T, Sjöros T, Laine S, Vähä-Ypyä H, Löyttyniemi E, 2010. DOI: 10.1210/jc.2009-1101.
Sievänen H, Houttu N, Laitinen K, Kalliokoski K, Vasankari T, Knuuti 115. Haus JM, Solomon TPJ, Marchetti CM, O’leary VB, Brooks LM,
J, Heinonen I. Effects of reduced sedentary time on cardiometabolic Gonzalez F, Kirwan JP. Decreased visfatin after exercise training
correlates with improved glucose tolerance. Med Sci Sports Exerc 41: study (DOPPS). Am J Kidney Dis Off J Natl Kidney Found 67: 283-292,
1255-1260, 2009. DOI: 10.1249/MSS.0b013e318195bad5. 2016. DOI: 10.1053/j.ajkd.2015.09.024.
116. Hawkins M, Tonelli J, Kishore P, Stein D, Ragucci E, Gitig A, Reddy 137. Jiwani SS, Carrillo-Larco RM, Hernández-Vásquez A, Barrientos-
K. Contribution of elevated free fatty acid levels to the lack of glucose Gutiérrez T, Basto-Abreu A, Gutierrez L, Irazola V, Nieto-Martínez
effectiveness in type 2 diabetes. Diabetes 52: 2748-2758, 2003. DOI: R, Nunes BP, Parra DC, Miranda JJ. The shift of obesity burden by
10.2337/diabetes.52.11.2748. socioeconomic status between 1998 and 2017 in Latin America and
117. Hawley JA, Lessard SJ. Exercise training-induced improvements in the Caribbean: A cross-sectional series study. Lancet Glob Health 7:
insulin action. Acta Physiol 192: 127-135, 2008. DOI: 10.1111/j.1748- e1644-e1654, 2019. DOI: 10.1016/S2214-109X(19)30421-8.
1716.2007.01783.x. 138. Johansen KL, Delgado C, Bao Y, Tamura MK. Frailty and dialysis ini-
118. He J, Watkins S, Kelley DE. Skeletal muscle lipid content and oxidative tiation. Semin Dial 26: 690-696, 2013. DOI: 10.1111/sdi.12126.
enzyme activity in relation to muscle fiber type in type 2 diabetes and 139. Johansen MY, MacDonald CS, Hansen KB, Karstoft K, Christensen
obesity. Diabetes 50: 817-823, 2001. DOI: 10.2337/diabetes.50.4.817. R, Pedersen M, Hansen LS, Zacho M, Wedell-Neergaard A-S, Nielsen
119. Heden TD, Winn NC, Mari A, Booth FW, Rector RS, Thyfault JP, ST, Iepsen UW, Langberg H, Vaag AA, Pedersen BK, Ried-Larsen
Kanaley JA. Postdinner resistance exercise improves postprandial M. Effect of an intensive lifestyle intervention on glycemic control
risk factors more effectively than predinner resistance exercise in in patients with type 2 diabetes. JAMA 318: 637-646, 2017. DOI:
patients with type 2 diabetes. J Appl Physiol 118: 624-634, 1985. DOI: 10.1001/jama.2017.10169.
10.1152/japplphysiol.00917.2014. 140. Jorge MLMP, de Oliveira VN, Resende NM, Paraiso LF, Calixto A,
120. Hellsten Y, Nyberg M. Cardiovascular adaptations to exercise training. Diniz ALD, Resende ES, Ropelle ER, Carvalheira JB, Espindola FS,
Compr Physiol 6, 2015. DOI: 10.1002/cphy.c140080. Jorge PT, Geloneze B. The effects of aerobic, resistance, and combined
121. Henson J, Edwardson CL, Davies MJ, Gill JMR, Heaney LM, Khunti exercise on metabolic control, inflammatory markers, adipocytokines,
K, Ng L, Sattar N, Zaccardi F, Yates T. Physical activity and lipidomics and muscle insulin signaling in patients with type 2 diabetes melli-
in a population at high risk of type 2 diabetes mellitus. J Sports Sci 38: tus. Metab Clin Exp 60: 1244-1252, 2011. DOI: 10.1016/j.metabol.
1150-1160, 2020. DOI: 10.1080/02640414.2020.1744836. 2011.01.006.
122. Holliday A, Blannin A. Appetite, food intake and gut hormone 141. Jung DY, Ko HJ, Lichtman EI, Lee E, Lawton E, Ong H, Yu K, Azuma
responses to intense aerobic exercise of different duration. J Endocrinol Y, Friedline RH, Lee KW, Kim JK. Short-term weight loss attenu-
235: 193-205, 2017. DOI: 10.1530/JOE-16-0570. ates local tissue inflammation and improves insulin sensitivity without
123. Holloszy JO. Biochemical adaptations in muscle: Effects of exercise affecting adipose inflammation in obese mice. Am J Physiol Endocrinol
on mitochondrial oxygen uptake and respiratory enzyme activ- Metab 304: E964-E976, 2013. DOI: 10.1152/ajpendo.00462.2012.
ity in skeletal muscle. J Biol Chem 242: 2278-2282, 1967. DOI: 142. Kahn CR. Insulin resistance, insulin insensitivity, and insulin unre-
10.1016/S0021-9258(18)96046-1. sponsiveness: A necessary distinction. Metabolism 27: 1893-1902,
124. Holloszy JO. Skeletal muscle “mitochondrial deficiency” does not 1978. DOI: 10.1016/S0026-0495(78)80007-9.
mediate insulin resistance. Am J Clin Nutr 89: 463S-466S, 2009. DOI: 143. Karstoft K, Clark MA, Jakobsen I, Knudsen SH, van Hall G, Pedersen
10.3945/ajcn.2008.26717C. BK, Solomon TPJ. Glucose effectiveness, but not insulin sensitivity,
125. Holten MK, Zacho M, Gaster M, Juel C, Wojtaszewski JFP, Dela F. is improved after short-term interval training in individuals with type 2
Strength training increases insulin-mediated glucose uptake, GLUT4 diabetes mellitus: A controlled, randomised, crossover trial. Diabetolo-
content, and insulin signaling in skeletal muscle in patients with type 2 gia 60: 2432-2442, 2017. DOI: 10.1007/s00125-017-4406-0.
diabetes. Diabetes 53: 294-305, 2004. DOI: 10.2337/diabetes.53.2.294. 144. Karstoft K, Wallis GA, Pedersen BK, Solomon TPJ. The effects
126. Hordern MD, Dunstan DW, Prins JB, Baker MK, Singh MAF, of interval- vs. continuous exercise on excess post-exercise oxygen
Coombes JS. Exercise prescription for patients with type 2 diabetes consumption and substrate oxidation rates in subjects with type 2 dia-
and pre-diabetes: A position statement from exercise and sport science betes. Metab Clin Exp 65: 1316-1325, 2016. DOI: 10.1016/j.metabol.
Australia. J Sci Med Sport 15: 25-31, 2012. DOI: 10.1016/j.jsams. 2016.05.017.
2011.04.005. 145. Kasumov T, Solomon TPJ, Hwang C, Huang H, Haus JM, Zhang R,
127. Hoshino J. Renal rehabilitation: Exercise intervention and nutri- Kirwan JP. Improved insulin sensitivity after exercise training is linked
tional support in dialysis patients. Nutrients 13: 1444, 2021. DOI: to reduced plasma C14:0 ceramide in obesity and type 2 diabetes.
10.3390/nu13051444. Obes Silver Spring Md 23: 1414-1421, 2015. DOI: 10.1002/oby.21117.
128. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegel- 146. Kawanishi N, Niihara H, Mizokami T, Yada K, Suzuki K. Exercise
man BM. IRS-1-Mediated inhibition of insulin receptor tyrosine kinase training attenuates neutrophil infiltration and elastase expression in
activity in TNF-α- and obesity-induced insulin resistance. Science 271: adipose tissue of high-fat-diet-induced obese mice. Physiol Rep 3:
665-670, 1996. DOI: 10.1126/science.271.5249.665. e12534, 2015. DOI: 10.14814/phy2.12534.
129. Houzelle A, Jörgensen JA, Schaart G, Daemen S, van Polanen N, 147. Kayaba M, Iwayama K, Ogata H, Seya Y, Kiyono K, Satoh M,
Fealy CE, Hesselink MKC, Schrauwen P, Hoeks J. Human skeletal Tokuyama K. The effect of nocturnal blue light exposure from
muscle mitochondrial dynamics in relation to oxidative capac- light-emitting diodes on wakefulness and energy metabolism the
ity and insulin sensitivity. Diabetologia 64: 424-436, 2021. DOI: following morning. Environ Health Prev Med 19: 354-361, 2014. DOI:
10.1007/s00125-020-05335-w. 10.1007/s12199-014-0402-x.
130. Howlett KF, Sakamoto K, Garnham A, Cameron-Smith D, Hargreaves 148. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mito-
M. Resistance exercise and insulin regulate AS160 and interaction chondria in human skeletal muscle in type 2 diabetes. Diabetes 51:
with 14-3-3 in human skeletal muscle. Diabetes 56: 1608-1614, 2007. 2944-2950, 2002. DOI: 10.2337/diabetes.51.10.2944.
DOI: 10.2337/db06-1398. 149. Kelly KR, Brooks LM, Solomon TPJ, Kashyap SR, O’Leary VB,
131. Huang M, Lv A, Wang J, Xu N, Ma G, Zhai Z, Zhang B, Gao J, Ni Kirwan JP. The glucose-dependent insulinotropic polypeptide and
C. Exercise training and outcomes in hemodialysis patients: System- glucose-stimulated insulin response to exercise training and diet in
atic review and meta-analysis. Am J Nephrol 50: 240-254, 2019. DOI: obesity. Am J Physiol Endocrinol Metab 296: E1269-E1274, 2009.
10.1159/000502447. DOI: 10.1152/ajpendo.00112.2009.
132. Huang X, Liu G, Guo J, Su Z. The PI3K/AKT pathway in obe- 150. Kelly KR, Haus JM, Solomon TPJ, Patrick-Melin AJ, Cook M, Rocco
sity and type 2 diabetes. Int J Biol Sci 14: 1483-1496, 2018. DOI: M, Barkoukis H, Kirwan JP. A low-glycemic index diet and exercise
10.7150/ijbs.27173. intervention reduces TNFα in isolated mononuclear cells of older,
133. Huffman KM, Koves TR, Hubal MJ, Abouassi H, Beri N, Bateman obese adults. J Nutr 141: 1089-1094, 2011. DOI: 10.3945/jn.111.
LA, Stevens RD, Ilkayeva OR, Hoffman EP, Muoio DM, Kraus WE. 139964.
Metabolite signatures of exercise training in human skeletal muscle 151. Kelly KR, Navaneethan SD, Solomon TPJ, Haus JM, Cook M,
relate to mitochondrial remodelling and cardiometabolic fitness. Dia- Barkoukis H, Kirwan JP. Lifestyle-induced decrease in fat mass
betologia 57: 2282-2295, 2014. DOI: 10.1007/s00125-014-3343-4. improves adiponectin secretion in obese adults. Med Sci Sports Exerc
134. Hundal RS, Krssak M, Dufour S, Laurent D, Lebon V, Chan- 46: 920-926, 2014. DOI: 10.1249/MSS.0000000000000200.
dramouli V, Inzucchi SE, Schumann WC, Petersen KF, Landau 152. Kennedy JM, Zochodne DW. Experimental diabetic neuropathy with
BR, Shulman GI. Mechanism by which metformin reduces glucose spontaneous recovery. Diabetes 54: 830-837, 2005.
production in type 2 diabetes. Diabetes 49: 2063-2069, 2000. DOI: 153. Khan MAB, Hashim MJ, King JK, Govender RD, Mustafa H, Al
10.2337/diabetes.49.12.2063. KJ. Epidemiology of type 2 diabetes – global burden of disease and
135. Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin forecasted trends. J Epidemiol Glob Health 10: 107-111, 2020. DOI:
resistance in human muscle is associated with changes in diacylglyc- 10.2991/jegh.k.191028.001.
erol, protein kinase C, and I𝜅B-α. Diabetes 51: 2005-2011, 2002. DOI: 154. Kim D-Y, Jung S-Y, Kim T-W, Lee K-S, Kim K. Treadmill exercise
10.2337/diabetes.51.7.2005. decreases incidence of Alzheimer’s disease by suppressing glycogen
136. Jassal SV, Karaboyas A, Comment LA, Bieber BA, Morgenstern synthase kinase-3β expression in streptozotocin-induced diabetic rats.
H, Sen A, Gillespie BW, De Sequera P, Marshall MR, Fukuhara J Exerc Rehabil 11: 87-94, 2015. DOI: 10.12965/jer.150198.
S, Robinson BM, Pisoni RL, Tentori F. Functional dependence and 155. Kim Y, Sharp S, Hwang S, Jee SH. Exercise and incidence of myocar-
mortality in the international dialysis outcomes and practice patterns dial infarction, stroke, hypertension, type 2 diabetes and site-specific
cancers: Prospective cohort study of 257 854 adults in South Korea. 176. Lee W, Almalki O. Recent advancements in bariatric/metabolic
BMJ Open 9: e025590, 2019. DOI: 10.1136/bmjopen-2018-025590. surgery. Ann Gastroenterol Surg 1: 171-179, 2017. DOI: 10.1002/ags3
156. Kim Y-B, Ciaraldi TP, Kong A, Kim D, Chu N, Mohideen P, Mudaliar .12030.
S, Henry RR, Kahn BB. Troglitazone but not Metformin restores 177. Lees AJ, Hardy J, Revesz T. Parkinson’s disease. Lancet 373: 2055-
insulin-stimulated phosphoinositide 3-kinase activity and increases 2066, 2009. DOI: 10.1016/S0140-6736(09)60492-X.
p110β protein levels in skeletal muscle of type 2 diabetic subjects. 178. Lenard NR, Berthoud H-R. Central and peripheral regulation of food
Diabetes 51: 443-448, 2002. intake and physical activity: Pathways and genes. Obesity 16: S11-S22,
157. Kirwan JP, Courcoulas AP, Cummings DE, Goldfine AB, Kashyap SR, 2008. DOI: 10.1038/oby.2008.511.
Simonson DC, Arterburn DE, Gourash WF, Vernon AH, Jakicic JM, 179. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell
Patti ME, Wolski K, Schauer PR. Diabetes remission in the alliance 132: 27-42, 2008. DOI: 10.1016/j.cell.2007.12.018.
of randomized trials of medicine versus metabolic surgery in type 2 180. Li D, Yang Y, Gao Z, Zhao L, Yang X, Xu F, Yu C, Zhang X, Wang
diabetes (ARMMS-T2D). Diabetes Care 45: 1574-1583, 2022. DOI: X, Wang L, Su J. Sedentary lifestyle and body composition in type 2
10.2337/dc21-2441. diabetes. Diabetol Metab Syndr 14: 8, 2022. DOI: 10.1186/s13098-
158. Kirwan JP, del Aguila LF, Hernandez JM, Williamson DL, O’Gorman 021-00778-6.
DJ, Lewis R, Krishnan RK. Regular exercise enhances insulin activa- 181. Li G, Zhang P, Wang J, An Y, Gong Q, Gregg EW, Yang W, Zhang B,
tion of IRS-1-associated PI3-kinase in human skeletal muscle. J Appl Shuai Y, Hong J, Engelgau MM, Li H, Roglic G, Hu Y, Bennett PH.
Physiol 88: 797-803, 2000. DOI: 10.1152/jappl.2000.88.2.797. Cardiovascular mortality, all-cause mortality, and diabetes incidence
159. Kirwan JP, Sacks J, NieuwoudtI S. The essential role of exercise in the after lifestyle intervention for people with impaired glucose tolerance
management of type 2 diabetes. Cleve Clin J Med 84: S15-S21, 2017. in the Da Qing Diabetes Prevention Study: A 23-year follow-up study.
DOI: 10.3949/ccjm.84.s1.03. Lancet Diabetes Endocrinol 2: 474-480, 2014. DOI: 10.1016/S2213-
160. Kirwan JP, Solomon TPJ, Wojta DM, Staten MA, Holloszy JO. Effects 8587(14)70057-9.
of 7 days of exercise training on insulin sensitivity and responsive- 182. Li J, Cheng W, Ma H. A comparative study of health efficacy indi-
ness in type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 297: cators in subjects with T2DM applying power cycling to 12 weeks
E151-E156, 2009. DOI: 10.1152/ajpendo.00210.2009. of low-volume high-intensity interval training and moderate-intensity
161. Klip A, McGraw TE, James DE. Thirty sweet years of GLUT4. J Biol continuous training. J Diabetes Res 2022: 9273830, 2022. DOI:
Chem 294: 11369-11381, 2019. DOI: 10.1074/jbc.REV119.008351. 10.1155/2022/9273830.
162. Klip A, Ramlal T, Young DA, Holloszy JO. Insulin-induced transloca- 183. Lin X, Zhang X, Guo J, Roberts CK, McKenzie S, Wu W-C, Liu S,
tion of glucose transporters in rat hindlimb muscles. FEBS Lett 224: Song Y. Effects of exercise training on cardiorespiratory fitness and
224-230, 1987. DOI: 10.1016/0014-5793(87)80452-0. biomarkers of cardiometabolic health: A systematic review and meta-
163. Kluding PM, Pasnoor M, Singh R, Jernigan S, Farmer K, Rucker J, analysis of randomized controlled trials. J Am Heart Assoc Cardiovasc
Sharma N, Wright DE. The effect of exercise on neuropathic symp- Cerebrovasc Dis 4: e002014, 2015. DOI: 10.1161/JAHA.115.002014.
toms, nerve function, and cutaneous innervation in people with diabetic 184. Linden MA, Fletcher JA, Morris EM, Meers GM, Kearney ML,
peripheral neuropathy. J Diabetes Complicat 26: 424-429, 2012. DOI: Crissey JM, Laughlin MH, Booth FW, Sowers JR, Ibdah JA, Thy-
10.1016/j.jdiacomp.2012.05.007. fault JP, Rector RS. Combining metformin and aerobic exercise
164. Konopka AR, Laurin JL, Schoenberg HM, Reid JJ, Castor WM, training in the treatment of type 2 diabetes and NAFLD in OLETF
Wolff CA, Musci RV, Safairad OD, Linden MA, Biela LM, Bailey rats. Am J Physiol Endocrinol Metab 306: E300-E310, 2014. DOI:
SM, Hamilton KL, Miller BF. Metformin inhibits mitochondrial 10.1152/ajpendo.00427.2013.
adaptations to aerobic exercise training in older adults. Aging Cell 18: 185. Ling C, Del Guerra S, Lupi R, Rönn T, Granhall C, Luthman H,
e12880, 2019. DOI: 10.1111/acel.12880. Masiello P, Marchetti P, Groop L, Del Prato S. Epigenetic regula-
165. Koo JH, Kwon IS, Kang EB, Lee CK, Lee NH, Kwon MG, Cho IH, tion of PPARGC1A in human type 2 diabetic islets and effect on
Cho J, yong. Neuroprotective effects of treadmill exercise on BDNF insulin secretion. Diabetologia 51: 615-622, 2008. DOI: 10.1007/
and PI3-K/Akt signaling pathway in the cortex of transgenic mice s00125-007-0916-5.
model of Alzheimer’s disease. J Exerc Nutr Biochem 17: 151-160, 186. Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell
2013. DOI: 10.5717/jenb.2013.17.4.151. Metab 29: 1028, 2019. DOI: 10.1016/j.cmet.2019.03.009.
166. Kramer A. An overview of the beneficial effects of exercise on health 187. Lowell BB, Shulman GI. Mitochondrial Dysfunction and Type 2 Dia-
and performance. In: Xiao J, editor. Physical Exercise for Human betes. Science 307: 384-387, 2005. DOI: 10.1126/science.1104343.
Health. Springer, p. 3-22. 188. Lund S, Holman GD, Schmitz O, Pedersen O. Contraction stimulates
167. Kraniou GN, Cameron-Smith D, Hargreaves M. Acute exercise and translocation of glucose transporter GLUT4 in skeletal muscle through
GLUT4 expression in human skeletal muscle: Influence of exercise a mechanism distinct from that of insulin. Proc Natl Acad Sci U S A 92:
intensity. J Appl Physiol 101: 934-937, 2006. DOI: 10.1152/japplphys- 5817-5821, 1995.
iol.01489.2005. 189. Maezawa Y, Takemoto M, Yokote K. Cell biology of diabetic
168. Ku Y, Han K, Ahn H, Kwon H, Koo B, Kim H, Min K. Resis- nephropathy: Roles of endothelial cells, tubulointerstitial cells and
tance exercise did not alter intramuscular adipose tissue but reduced podocytes. J Diabetes Investig 6: 3-15, 2015. DOI: 10.1111/jdi.12255.
retinol-binding protein-4 concentration in individuals with type 2 190. Mahdi A, Jiao T, Yang J, Kövamees O, Alvarsson M, von Heijne M,
diabetes mellitus. J Int Med Res 38: 782-791, 2010. DOI: 10.1177/ Zhou Z, Pernow J. The effect of glycemic control on endothelial and
147323001003800305. cardiac dysfunction induced by red blood cells in type 2 diabetes. Front
169. Kullman EL, Kelly KR, Haus JM, Fealy CE, Scelsi AR, Pagadala Pharmacol 10: 861, 2019. DOI: 10.3389/fphar.2019.00861.
MR, Flask CA, McCullough AJ, Kirwan JP. Short-term aerobic 191. Maiorana A, O’Driscoll G, Cheetham C, Dembo L, Stanton K,
exercise training improves gut peptide regulation in nonalcoholic Goodman C, Taylor R, Green D. The effect of combined aero-
fatty liver disease. J Appl Physiol 120: 1159-1164, 2016. DOI: bic and resistance exercise training on vascular function in type 2
10.1152/japplphysiol.00693.2015. diabetes. J Am Coll Cardiol 38: 860-866, 2001. DOI: 10.1016/S0735-
170. Lancaster GI, Febbraio MA. The immunomodulating role of exercise 1097(01)01439-5.
in metabolic disease. Trends Immunol 35: 262-269, 2014. DOI: 192. Malin SK, Gerber R, Chipkin SR, Braun B. Independent and com-
10.1016/j.it.2014.02.008. bined effects of exercise training and metformin on insulin sensitivity
171. Latres E, Amini AR, Amini AA, Griffiths J, Martin FJ, Wei Y, in individuals with prediabetes. Diabetes Care 35: 131-136, 2012. DOI:
Lin HC, Yancopoulos GD, Glass DJ. Insulin-like growth factor-1 10.2337/dc11-0925.
(IGF-1) inversely regulates atrophy-induced genes via the phos- 193. Malin SK, Rincon JPD, Huang H, Kirwan JP. Exercise-induced lower-
phatidylinositol 3-kinase/Akt/mammalian target of rapamycin ing of fetuin-A may increase hepatic insulin sensitivity. Med Sci Sports
(PI3K/Akt/mTOR) pathway. J Biol Chem 280: 2737-2744, 2005. DOI: Exerc 46: 2085-2090, 2014. DOI: 10.1249/MSS.0000000000000338.
10.1074/jbc.M407517200. 194. Malin SK, Solomon TPJ, Blaszczak A, Finnegan S, Filion J, Kirwan
172. Lawrence RD. The effect of exercise on insulin action in diabetes. JP. Pancreatic β-cell function increases in a linear dose-response
Br Med J 1: 648-650, 1926. manner following exercise training in adults with prediabetes.
173. Lecker SH, Goldberg AL, Mitch WE. Protein degradation by the Am J Physiol Endocrinol Metab 305: E1248-E1254, 2013. DOI:
ubiquitin–proteasome pathway in normal and disease states. J Am Soc 10.1152/ajpendo.00260.2013.
Nephrol 17: 1807-1819, 2006. DOI: 10.1681/ASN.2006010083. 195. Mann TN, Webster C, Lamberts RP, Lambert MI. Effect of exercise
174. Lee J-H, Lee R, Hwang M-H, Hamilton MT, Park Y. The effects of intensity on post-exercise oxygen consumption and heart rate recovery.
exercise on vascular endothelial function in type 2 diabetes: a system- Eur J Appl Physiol 114: 1809-1820, 2014. DOI: 10.1007/s00421-014-
atic review and meta-analysis. Diabetol Metab Syndr 10: 15, 2018. 2907-9.
DOI: 10.1186/s13098-018-0316-7. 196. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JFE,
175. Lee SS, Yoo JH, So YS. Effect of the low- versus high-intensity Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg
exercise training on endoplasmic reticulum stress and GLP-1 in WM, Stockner M, Zinman B, Bergenstal RM, Buse JB. Liraglutide
adolescents with type 2 diabetes mellitus. J Phys Ther Sci 27: 3063, and cardiovascular outcomes in type 2 diabetes. N Engl J Med 375:
2015. DOI: 10.1589/jpts.27.3063. 311-322, 2016. DOI: 10.1056/NEJMoa1603827.
197. Martins C, Kulseng B, Rehfeld JF, King NA, Blundell JE. Effect of kinase activity in skeletal muscle of subjects with type 2 diabetes.
chronic exercise on appetite control in overweight and obese individ- Diabetes 51: 2074-2081, 2002. DOI: 10.2337/diabetes.51.7.2074.
uals. Med Sci Sports Exerc 45: 805-812, 2013. DOI: 10.1249/MSS. 214. Myette-Côté É, Terada T, Boulé NG. The effect of exercise with or
0b013e31827d1618. without metformin on glucose profiles in type 2 diabetes: A pilot study.
198. Martins C, Stensvold D, Finlayson G, Holst J, Wisloff U, Kulseng Can J Diabetes 40: 173-177, 2016. DOI: 10.1016/j.jcjd.2015.08.015.
B, Morgan L, King NA. Effect of moderate- and high-intensity acute 215. Nathan DM, Barrett-Connor E, Crandall JP, Edelstein SL, Goldberg
exercise on appetite in obese individuals. Med Sci Sports Exerc 47: RB, Horton ES, Knowler WC, Mather KJ, Orchard TJ, Pi-Sunyer X,
40-48, 2015. DOI: 10.1249/MSS.0000000000000372. Schade D, Temprosa M. Long-term effects of lifestyle intervention
199. Maser RE, Mitchell BD, Vinik AI, Freeman R. The association between or metformin on diabetes development and microvascular compli-
cardiovascular autonomic neuropathy and mortality in individuals with cations: The DPP outcomes study. Lancet Diabetes Endocrinol 3:
diabetes: A meta-analysis. Diabetes Care 26: 1895-1901, 2003. DOI: 866-875, 2015. DOI: 10.1016/S2213-8587(15)00291-0.
10.2337/diacare.26.6.1895. 216. National Institute of Health. Molecular transducers of physical activ-
200. Matos VAF, Souza DC, Santos VOA, Medeiros ÍF, Browne RAV, ity in humans - Overview [Online], 2015. https://commonfund.nih.gov/
Nascimento PRP, Marinho CSR, Serquiz AC, Costa EC, Fayh APT. moleculartransducers/overview [29 Aug. 2021].
Acute effects of high-intensity interval and moderate-intensity contin- 217. Nauck M. Incretin therapies: Highlighting common features and dif-
uous exercise on GLP-1, appetite and energy intake in obese men: A ferences in the modes of action of glucagon-like peptide-1 receptor
crossover trial. Nutrients 10: 889, 2018. DOI: 10.3390/nu10070889. agonists and dipeptidyl peptidase-4 inhibitors. Diabetes Obes Metab
201. Mavros Y, Kay S, Simpson KA, Baker MK, Wang Y, Zhao RR, Meik- 18: 203-216, 2016. DOI: 10.1111/dom.12591.
lejohn J, Climstein M, O’Sullivan AJ, de Vos N, Baune BT, Blair SN, 218. Navaneethan SD, Fealy CE, Scelsi A, Arrigain S, Malin SK, Kirwan
Simar D, Rooney K, Singh NA, Fiatarone Singh MA. Reductions in JP. A trial of lifestyle modification on cardiopulmonary, inflammatory,
C-reactive protein in older adults with type 2 diabetes are related to and metabolic effects among obese with chronic kidney disease. Am J
improvements in body composition following a randomized controlled Nephrol 42: 274-281, 2015. DOI: 10.1159/000441155.
trial of resistance training. J Cachexia Sarcopenia Muscle 5: 111-120, 219. Nieuwoudt S, Fealy CE, Foucher JA, Scelsi AR, Malin SK, Pagadala
2014. DOI: 10.1007/s13539-014-0134-1. M, Rocco M, Burguera B, Kirwan JP. Functional high-intensity
202. McFarlin BK, Flynn MG, Campbell WW, Craig BA, Robinson JP, training improves pancreatic β-cell function in adults with type 2
Stewart LK, Timmerman KL, Coen PM. Physical activity status, but diabetes. Am J Physiol Endocrinol Metab 313: E314-E320, 2017.
not age, influences inflammatory biomarkers and toll-like receptor 4. DOI: 10.1152/ajpendo.00407.2016.
J Gerontol Ser A 61: 388-393, 2006. DOI: 10.1093/gerona/61.4.388. 220. Nieuwoudt S, Mulya A, Fealy CE, Martelli E, Dasarathy S, Naga Prasad
203. Meex RCR, Schrauwen-Hinderling VB, Moonen-Kornips E, Schaart SV, Kirwan JP. In vitro contraction protects against palmitate-induced
G, Mensink M, Phielix E, van de Weijer T, Sels J-P, Schrauwen P, insulin resistance in C2C12 myotubes. Am J Physiol Cell Physiol 313:
Hesselink MKC. Restoration of muscle mitochondrial function and C575-C583, 2017. DOI: 10.1152/ajpcell.00123.2017.
metabolic flexibility in type 2 diabetes by exercise training is paralleled 221. O’Gorman DJ, Del Aguila LF, Williamson DL, Krishnan RK, Kirwan
by increased myocellular fat storage and improved insulin sensitivity. JP. Insulin and exercise differentially regulate PI3-kinase and glyco-
Diabetes 59: 572-579, 2009. DOI: 10.2337/db09-1322. gen synthase in human skeletal muscle. J Appl Physiol 89: 1412-1419,
204. Mensberg P, Nyby S, Jørgensen PG, Storgaard H, Jensen MT, 2000. DOI: 10.1152/jappl.2000.89.4.1412.
Sivertsen J, Holst JJ, Kiens B, Richter EA, Knop FK, Vilsbøll T. 222. Okada S, Hiuge A, Makino H, Nagumo A, Takaki H, Konishi H,
Near-normalization of glycaemic control with glucagon-like peptide-1 Goto Y, Yoshimasa Y, Miyamoto Y. Effect of exercise intervention on
receptor agonist treatment combined with exercise in patients with endothelial function and incidence of cardiovascular disease in patients
type 2 diabetes. Diabetes Obes Metab 19: 172-180, 2017. DOI: with type 2 diabetes. J Atheroscler Thromb 17: 828-833, 2010. DOI:
10.1111/dom.12797. 10.5551/jat.3798.
205. Misra A, Alappan NK, Vikram NK, Goel K, Gupta N, Mittal K, Bhatt S, 223. Oliveira AG, Araujo TG, Carvalho BM, Guadagnini D, Rocha GZ,
Luthra K. Effect of supervised progressive resistance-exercise training Bagarolli RA, Carvalheira JBC, Saad MJA. Acute exercise induces
protocol on insulin sensitivity, glycemia, lipids, and body composition a phenotypic switch in adipose tissue macrophage polarization in
in asian indians with type 2 diabetes. Diabetes Care 31: 1282-1287, diet-induced obese rats. Obesity 21: 2545-2556, 2013. DOI: 10.1002/
2008. DOI: 10.2337/dc07-2316. oby.20402.
206. Mogensen M, Sahlin K, Fernström M, Glintborg D, Vind BF, Beck- 224. Orru H, Ebi KL, Forsberg B. The interplay of climate change and air
Nielsen H, Højlund K. Mitochondrial respiration is decreased in skele- pollution on health. Curr Environ Health Rep 4: 504-513, 2017. DOI:
tal muscle of patients with type 2 diabetes. Diabetes 56: 1592-1599, 10.1007/s40572-017-0168-6.
2007. DOI: 10.2337/db06-0981. 225. Osborn O, Olefsky JM. The cellular and signaling networks linking
207. Montvida O, Shaw J, Atherton JJ, Stringer F, Paul SK. Long-term the immune system and metabolism in disease. Nat Med 18: 363-374,
trends in antidiabetes drug usage in the US: Real-world evidence in 2012. DOI: 10.1038/nm.2627.
patients newly diagnosed with type 2 diabetes. Diabetes Care 41: 226. Ostrowski K, Schjerling P, Pedersen BK. Physical activity and plasma
69-78, 2018. DOI: 10.2337/dc17-1414. interleukin-6 in humans – Effect of intensity of exercise. Eur J Appl
208. Moran C, Phan TG, Chen J, Blizzard L, Beare R, Venn A, Münch G, Physiol 83: 512-515, 2000. DOI: 10.1007/s004210000312.
Wood AG, Forbes J, Greenaway TM, Pearson S, Srikanth V. Brain 227. Pan DA, Lillioja S, Kriketos AD, Milner MR, Baur LA, Bogardus
atrophy in type 2 diabetes. Diabetes Care 36: 4036-4042, 2013. DOI: C, Jenkins AB, Storlien LH. Skeletal muscle triglyceride levels are
10.2337/dc13-0143. inversely related to insulin action. Diabetes 46: 983-988, 1997. DOI:
209. Mori K. Maintenance of skeletal muscle to counteract sarcopenia 10.2337/diab.46.6.983.
in patients with advanced chronic kidney disease and especially 228. Park S, Zachary WW, Gittelsohn J, Quinn CC, Surkan PJ. Neighbor-
those undergoing hemodialysis. Nutrients 13: 1538, 2021. DOI: hood influences on physical activity among low-income African Amer-
10.3390/nu13051538. ican adults with type 2 diabetes mellitus. Diabetes Educ 46: 181-190,
210. Motiani KK, Collado MC, Eskelinen J-J, Virtanen KA, Loyttyniemi 2020. DOI: 10.1177/0145721720906082.
E, Salminen S, Nuutila P, Kalliokoski KK, Hannukainen JC. Exercise 229. Parry SA, Hodson L. Managing NAFLD in type 2 diabetes: The effect
training modulates gut microbiota profile and improves endotox- of lifestyle interventions, a narrative review. Adv Ther 37: 1381-1406,
emia. Med Sci Sports Exerc 52: 94-104, 2020. DOI: 10.1249/MSS. 2020. DOI: 10.1007/s12325-020-01281-6.
0000000000002112. 230. Pasini E, Corsetti G, Assanelli D, Testa C, Romano C, Dioguardi FS,
211. Motiani KK, Savolainen AM, Toivanen J, Eskelinen J-J, Yli- Aquilani R. Effects of chronic exercise on gut microbiota and intestinal
Karjanmaa M, Virtanen KA, Saunavaara V, Heiskanen MA, Parkkola barrier in human with type 2 diabetes. Minerva Med 110: 3-11, 2019.
R, Haaparanta-Solin M, Solin O, Savisto N, Löyttyniemi E, Knuuti DOI: 10.23736/S0026-4806.18.05589-1.
J, Nuutila P, Kalliokoski KK, Hannukainen JC. Effects of short- 231. Perry BD, Caldow MK, Brennan-Speranza TC, Sbaraglia M, Jerums G,
term sprint interval and moderate-intensity continuous training Garnham A, Wong C, Levinger P, Asrar ul Haq M, Hare DL, Price SR,
on liver fat content, lipoprotein profile, and substrate uptake: A Levinger I. Muscle atrophy in patients with type 2 diabetes mellitus:
randomized trial. J Appl Physiol 126: 1756-1768, 2019. DOI: Roles of inflammatory pathways, physical activity and exercise. Exerc
10.1152/japplphysiol.00900.2018. Immunol Rev 22: 94-109, 2016.
212. Murai IH, Roschel H, Dantas WS, Gil S, Merege-Filho C, de Cleva R, 232. Peters TM, Brazeau A-S. Exercise in pregnant women with diabetes.
de Sá-Pinto AL, Lima F, Santo MA, Benatti FB, Kirwan JP, Pereira Curr Diab Rep 19: 80, 2019. DOI: 10.1007/s11892-019-1204-8.
RM, Gualano B. Exercise mitigates bone loss in women with severe 233. Phielix E, Meex R, Moonen-Kornips E, Hesselink MKC, Schrauwen
obesity after roux-en-y gastric bypass: A randomized controlled trial. P. Exercise training increases mitochondrial content and ex vivo
J Clin Endocrinol Metab 104: 4639-4650, 2019. DOI: 10.1210/jc.2019- mitochondrial function similarly in patients with type 2 diabetes
00074. and in control individuals. Diabetologia 53: 1714-1721, 2010. DOI:
213. Musi N, Hirshman MF, Nygren J, Svanfeldt M, Bavenholm P, Rooy- 10.1007/s00125-010-1764-2.
ackers O, Zhou G, Williamson JM, Ljunqvist O, Efendic S, Moller DE, 234. Plomgaard P, Nielsen AR, Fischer CP, Mortensen OH, Broholm C,
Thorell A, Goodyear LJ. Metformin increases AMP-activated protein Penkowa M, Krogh-Madsen R, Erikstrup C, Lindegaard B, Petersen
AMW, Taudorf S, Pedersen BK. Associations between insulin resis- 254. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh
tance and TNF-α in plasma, skeletal muscle and adipose tissue in K, Schiaffino S, Lecker SH, Goldberg AL. Foxo transcription factors
humans with and without type 2 diabetes. Diabetologia 50: 2562-2571, induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal
2007. DOI: 10.1007/s00125-007-0834-6. muscle atrophy. Cell 117: 399-412, 2004.
235. Ploug T, Wojtaszewski J, Kristiansen S, Hespel P, Galbo H, Richter EA. 255. Santry HP, Gillen DL, Lauderdale DS. Trends in bariatric surgical
Glucose transport and transporters in muscle giant vesicles: Differential procedures. JAMA 294: 1909-1917, 2005. DOI: 10.1001/jama.294.
effects of insulin and contractions. Am J Phys 264: E270-E278, 1993. 15.1909.
DOI: 10.1152/ajpendo.1993.264.2.E270. 256. Sato Y. Diabetes and life-styles: Role of physical exercise for pri-
236. Poehlman ET, Melby C. Resistance training and energy balance. Int J mary prevention. Br J Nutr 84: S187-S190, 2000. DOI: 10.1079/
Sports Nutr 8: 143-159, 1998. 09658219738866.
237. Pop-Busui R. Cardiac autonomic neuropathy in diabetes. Diabetes 257. Savikj M, Gabriel BM, Alm PS, Smith J, Caidahl K, Björnholm M,
Care 33: 434-441, 2010. DOI: 10.2337/dc09-1294. Fritz T, Krook A, Zierath JR, Wallberg-Henriksson H. Afternoon
238. Potes Y, de Luxán-Delgado B, Rodriguez-González S, Guimarães exercise is more efficacious than morning exercise at improving
MRM, Solano JJ, Fernández-Fernández M, Bermúdez M, Boga JA, blood glucose levels in individuals with type 2 diabetes: A randomised
Vega-Naredo I, Coto-Montes A. Overweight in elderly people induces crossover trial. Diabetologia 62: 233-237, 2019. DOI: 10.1007/s00125-
impaired autophagy in skeletal muscle. Free Radic Biol Med 110: 018-4767-z.
31-41, 2017. DOI: 10.1016/j.freeradbiomed.2017.05.018. 258. Sawacha Z, Gabriella G, Cristoferi G, Guiotto A, Avogaro A, Cobelli
239. Prompers L, Schaper N, Apelqvist J, Edmonds M, Jude E, Mauricio C. Diabetic gait and posture abnormalities: A biomechanical investiga-
D, Uccioli L, Urbancic V, Bakker K, Holstein P, Jirkovska A, Piag- tion through three dimensional gait analysis. Clin Biomech 24: 722-728,
gesi A, Ragnarson-Tennvall G, Reike H, Spraul M, Van Acker K, Van 2009. DOI: 10.1016/j.clinbiomech.2009.07.007.
Baal J, Van Merode F, Ferreira I, Huijberts M. Prediction of outcome in 259. Schauer PR, Bhatt DL, Kirwan JP, Wolski K, Aminian A, Brethauer
individuals with diabetic foot ulcers: focus on the differences between SA, Navaneethan SD, Singh RP, Pothier CE, Nissen SE, Kashyap SR.
individuals with and without peripheral arterial disease: The EURODI- Bariatric surgery versus intensive medical therapy for diabetes — 5-
ALE Study. Diabetologia 51: 747-755, 2008. DOI: 10.1007/s00125- Year outcomes. N Engl J Med 376: 641-651, 2017. DOI: 10.1056/NEJ-
008-0940-0. Moa1600869.
240. Pugliese G, Penno G, Natali A, Barutta F, Di Paolo S, Reboldi G, 260. Schauer PR, Bhatt DL, Kirwan JP, Wolski K, Brethauer SA, Nava-
Gesualdo L, De Nicola L. Diabetic kidney disease: New clinical and neethan SD, Aminian A, Pothier CE, Kim ESH, Nissen SE, Kashyap
therapeutic issues. Joint position statement of the Italian Diabetes SR. Bariatric surgery versus intensive medical therapy for dia-
Society and the Italian Society of Nephrology on “The natural history betes – 3-Year outcomes. N Engl J Med 370: 2002-2013, 2014. DOI:
of diabetic kidney disease and treatment of hyperglycemia in patients 10.1056/NEJMoa1401329.
with type 2 diabetes and impaired renal function.”. J Nephrol 33: 9-35, 261. Schauer PR, Kashyap SR, Wolski K, Brethauer SA, Kirwan JP, Pothier
2020. DOI: 10.1007/s40620-019-00650-x. CE, Thomas S, Abood B, Nissen SE, Bhatt DL. Bariatric surgery versus
241. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, intensive medical therapy in obese patients with diabetes. N Engl J Med
Shen D, Peng Y, Zhang D, Jie Z, Wu W, Qin Y, Xue W, Li J, Han L, 366: 1567-1576, 2012. DOI: 10.1056/NEJMoa1200225.
Lu D, Wu P, Dai Y, Sun X, Li Z, Tang A, Zhong S, Li X, Chen W, 262. Scheele C, Nielsen AR, Walden TB, Sewell DA, Fischer CP, Brogan
Xu R, Wang M, Feng Q, Gong M, Yu J, Zhang Y, Zhang M, Hansen RJ, Petrovic N, Larsson O, Tesch PA, Wennmalm K, Hutchinson DS,
T, Sanchez G, Raes J, Falony G, Okuda S, Almeida M, LeChatelier E, Cannon B, Wahlestedt C, Pedersen BK, Timmons JA. Altered regula-
Renault P, Pons N, Batto J-M, Zhang Z, Chen H, Yang R, Zheng W, tion of the PINK1 locus: A link between type 2 diabetes and neurode-
Li S, Yang H, Wang J, Ehrlich SD, Nielsen R, Pedersen O, Kristiansen generation? FASEB J Off Publ Fed Am Soc Exp Biol 21: 3653-3665,
K, Wang J. A metagenome-wide association study of gut microbiota in 2007. DOI: 10.1096/fj.07-8520com.
type 2 diabetes. Nature 490: 55-60, 2012. DOI: 10.1038/nature11450. 263. Seuring T, Archangelidi O, Suhrcke M. The economic costs of type 2
242. Ren C, Liu W, Li J, Cao Y, Xu J, Lu P. Physical activity and risk of diabetes: A global systematic review. PharmacoEconomics 33: 811-
diabetic retinopathy: A systematic review and meta-analysis. Acta Dia- 831, 2015. DOI: 10.1007/s40273-015-0268-9.
betol 56: 823-837, 2019. DOI: 10.1007/s00592-019-01319-4. 264. Shah AS, El Ghormli L, Vajravelu ME, Bacha F, Farrell RM, Gidding
243. Richter EA, Garetto LP, Goodman MN, Ruderman NB. Muscle glu- SS, Levitt Katz LE, Tryggestad JB, White NH, Urbina EM. Heart
cose metabolism following exercise in the rat: Increased sensitivity to rate variability and cardiac autonomic dysfunction: prevalence, risk
insulin. J Clin Invest 69: 785-793, 1982. DOI: 10.1172/JCI110517. factors, and relationship to arterial stiffness in the treatment options
244. Richter EA, Hargreaves M. Exercise, GLUT4, and skeletal muscle for type 2 diabetes in adolescents and youth (TODAY) study. Diabetes
glucose uptake. Physiol Rev 93: 993-1017, 2013. DOI: 10.1152/phys- Care 42: 2143-2150, 2019. DOI: 10.2337/dc19-0993.
rev.00038.2012. 265. Shah M, Snell PG, Rao S, Adams-Huet B, Quittner C, Livingston
245. Riis S, Christensen B, Nellemann B, Møller AB, Husted AS, Pedersen EH, Garg A. High-volume exercise program in obese bariatric surgery
SB, Schwartz TW, Jørgensen JOL, Jessen N. Molecular adaptations patients: A randomized controlled trial. Obesity 19: 1826-1834, 2011.
in human subcutaneous adipose tissue after ten weeks of endurance DOI: 10.1038/oby.2011.172.
exercise training in healthy males. J Appl Physiol 126: 569-577, 2019. 266. Shakher J, Stevens MJ. Update on the management of diabetic polyneu-
DOI: 10.1152/japplphysiol.00989.2018. ropathies. Diabetes Metab Syndr Obes Targets Ther 4: 289-305, 2011.
246. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster BH, Kelley DOI: 10.2147/DMSO.S11324.
DE. Deficiency of subsarcolemmal mitochondria in obesity and type 2 267. Sheng K, Zhang P, Chen L, Cheng J, Wu C, Chen J. Intradialytic
diabetes. Diabetes 54: 8-14, 2005. DOI: 10.2337/diabetes.54.1.8. exercise in hemodialysis patients: A systematic review and meta-
247. Romijn JA, Coyle EF, Sidossis LS, Gastaldelli A, Horowitz JF, analysis. Am J Nephrol 40: 478-490, 2014. DOI: 10.1159/000368722.
Endert E, Wolfe RR. Regulation of endogenous fat and carbohydrate 268. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance.
metabolism in relation to exercise intensity and duration. Am J Physiol J Clin Invest 116: 1793-1801, 2006. DOI: 10.1172/JCI29069.
Endocrinol Metab 265: E380-E391, 1993. DOI: 10.1152/ajpendo. 269. Sigal RJ, Kenny GP, Boulé NG, Wells GA, Prud’homme D, Fortier M,
1993.265.3.E380. Reid RD, Tulloch H, Coyle D, Phillips P, Jennings A, Jaffey J. Effects of
248. Ryvicker M, Sridharan S. Neighborhood environment and disparities aerobic training, resistance training, or both on glycemic control in type
in health care access among urban medicare beneficiaries with diabetes: 2 diabetes. Ann Intern Med 147: 357-369, 2007. DOI: 10.7326/0003-
A retrospective cohort study. Inq J Health Care Organ Provis Financ 4819-147-6-200709180-00005.
55: 0046958018771414, 2018. DOI: 10.1177/0046958018771414. 270. Simpson RJ, Kunz H, Agha N, Graff R. Chapter Fifteen - Exercise and
249. Sadagurski M, Cheng Z, Rozzo A, Palazzolo I, Kelley GR, Dong X, the regulation of immune functions. In: Bouchard C, editor. Progress
Krainc D, White MF. IRS2 increases mitochondrial dysfunction and in Molecular Biology and Translational Science. Academic Press,
oxidative stress in a mouse model of Huntington disease. J Clin Invest p. 355-380.
121: 4070-4081, 2011. DOI: 10.1172/JCI46305. 271. Singleton JR, Smith AG, Marcus RL. Exercise as therapy for diabetic
250. Sadanand S, Balachandar R, Bharath S. Memory and executive func- and prediabetic neuropathy. Curr Diab Rep 15: 120, 2015. DOI:
tions in persons with type 2 diabetes: A meta-analysis. Diabetes Metab 10.1007/s11892-015-0682-6.
Res Rev 32: 132-142, 2016. DOI: 10.1002/dmrr.2664. 272. Singleton JR, Smith AG, Russell JW, Feldman EL. Microvascular
251. Sale DG, McComas AJ, MacDougall JD, Upton AR. Neuromuscular complications of impaired glucose tolerance. Diabetes 52: 2867-2873,
adaptation in human thenar muscles following strength training 2003.
and immobilization. J Appl Physiol 53: 419-424, 1982. DOI: 273. Smith AD, Crippa A, Woodcock J, Brage S. Physical activity and inci-
10.1152/jappl.1982.53.2.419. dent type 2 diabetes mellitus: a systematic review and dose–response
252. Saltiel AR. Insulin signaling in health and disease. J Clin Invest 131: meta-analysis of prospective cohort studies. Diabetologia 59: 2527-
e142241, 2021. DOI: 10.1172/JCI142241. 2545, 2016. DOI: 10.1007/s00125-016-4079-0.
253. Samsu N. Diabetic nephropathy: Challenges in pathogenesis, diag- 274. Smith-White MA, Wallace D, Potter EK. Sympathetic–parasympathetic
nosis, and treatment. Biomed Res Int 2021: 1497449, 2021. DOI: interactions at the heart in the anaesthetised rat. J Auton Nerv Syst 75:
10.1155/2021/1497449. 171-175, 1999. DOI: 10.1016/S0165-1838(98)00169-6.
275. Solomon TP, Haus JM, Kelly KR, Cook MD, Filion J, Rocco M, 293. Talbot K, Wang H-Y, Kazi H, Han L-Y, Bakshi KP, Stucky A, Fuino
Kashyap SR, Watanabe RM, Barkoukis H, Kirwan JP. A low–glycemic RL, Kawaguchi KR, Samoyedny AJ, Wilson RS, Arvanitakis Z,
index diet combined with exercise reduces insulin resistance, post- Schneider JA, Wolf BA, Bennett DA, Trojanowski JQ, Arnold SE.
prandial hyperinsulinemia, and glucose-dependent insulinotropic Demonstrated brain insulin resistance in Alzheimer’s disease patients
polypeptide responses in obese, prediabetic humans1234. Am J Clin is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive
Nutr 92: 1359-1368, 2010. DOI: 10.3945/ajcn.2010.29771. decline. J Clin Invest 122: 1316-1338, 2012. DOI: 10.1172/JCI59903.
276. Solomon TPJ, Haus JM, Kelly KR, Rocco M, Kashyap SR, Kirwan 294. Terauchi Y, Takada T, Yoshida S. A randomized controlled trial of
JP. Improved pancreatic β-cell function in type 2 diabetic patients a structured program combining aerobic and resistance exercise for
after lifestyle-induced weight loss is related to glucose-dependent adults with type 2 diabetes in Japan. Diabetol Int 13: 75-84, 2022.
insulinotropic polypeptide. Diabetes Care 33: 1561-1566, 2010. DOI: DOI: 10.1007/s13340-021-00506-5.
10.2337/dc09-2021. 295. Tesfaye S, Boulton AJM, Dyck PJ, Freeman R, Horowitz M, Kempler
277. Solomon TPJ, Malin SK, Karstoft K, Kashyap SR, Haus JM, Kirwan P, Lauria G, Malik RA, Spallone V, Vinik A, Bernardi L, Valensi P.
JP. Pancreatic β-cell function is a stronger predictor of changes in Diabetic neuropathies: Update on definitions, diagnostic criteria, esti-
glycemic control after an aerobic exercise intervention than insulin mation of severity, and treatments. Diabetes Care 33: 2285-2293, 2010.
sensitivity. J Clin Endocrinol Metab 98: 4176-4186, 2013. DOI: DOI: 10.2337/dc10-1303.
10.1210/jc.2013-2232. 296. The Diabetes Prevention Program Research Group. Reduction in the
278. Solomon TPJ, Malin SK, Karstoft K, Knudsen SH, Haus JM, Laye MJ, incidence of type 2 diabeteswith lifestyle intervention or Metformin.
Kirwan JP. Association between cardiorespiratory fitness and the deter- N Engl J Med 346: 393-403, 2002. DOI: 10.1056/NEJMoa012512.
minants of glycemic control across the entire glucose tolerance contin- 297. The Diabetes Prevention Program Research Group. Impact of intensive
uum. Diabetes Care 38: 921-929, 2015. DOI: 10.2337/dc14-2813. lifestyle and metformin therapy on cardiovascular disease risk factors
279. Spranger J, Kroke A, Möhlig M, Hoffmann K, Bergmann MM, in the diabetes prevention program. Diabetes Care 28: 888-894, 2005.
Ristow M, Boeing H, Pfeiffer AFH. Inflammatory cytokines and 298. The LookAHEAD Research Group. Cardiovascular effects of intensive
the risk to develop type 2 diabetes: Results of the prospective lifestyle intervention in type 2 diabetes. N Engl J Med 369: 145-154,
population-based European Prospective Investigation into Cancer and 2013. DOI: 10.1056/NEJMoa1212914.
Nutrition (EPIC)-Potsdam Study. Diabetes 52: 812-817, 2003. DOI: 299. The LookAHEAD Study Group, Gregg EW. Association of the mag-
10.2337/diabetes.52.3.812. nitude of weight loss and physical fitness change on long-term CVD
280. Sriwijitkamol A, Christ-Roberts C, Berria R, Eagan P, Pratipanawatr outcomes: The look AHEAD study. Lancet Diabetes Endocrinol 4:
T, DeFronzo RA, Mandarino LJ, Musi N. Reduced skeletal muscle 913-921, 2016. DOI: 10.1016/S2213-8587(16)30162-0.
inhibitor of 𝜅Bβ content is associated with insulin resistance in sub- 300. Thiering E, Heinrich J. Epidemiology of air pollution and diabetes.
jects with type 2 diabetes: Reversal by exercise training. Diabetes 55: Trends Endocrinol Metab 26: 384-394, 2015. DOI: 10.1016/j.tem.2015.
760-767, 2006. DOI: 10.2337/diabetes.55.03.06.db05-0677. 05.002.
281. Stack AG, Molony DA, Rives T, Tyson J, Murthy BVR. Association 301. Thomas PK. Classification, differential diagnosis, and staging of dia-
of physical activity with mortality in the US dialysis population. betic peripheral neuropathy. Diabetes 46 (Suppl 2): S54-S57, 1997.
Am J Kidney Dis Off J Natl Kidney Found 45: 690-701, 2005. DOI: DOI: 10.2337/diab.46.2.s54.
10.1053/j.ajkd.2004.12.013. 302. Tjønna AE, Lee SJ, Rognmo Ø, Stølen T, Bye A, Haram PM, Loen-
282. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. nechen JP, Al-Share QY, Skogvoll E, Slørdahl SA, Kemi OJ, Najjar
Exercise and IL-6 infusion inhibit endotoxin-induced TNF-α produc- SM, Wisløff U. Aerobic interval training vs. continuous moderate
tion in humans. FASEB J 17: 1-10, 2003. DOI: 10.1096/fj.02-0670fje. exercise as a treatment for the metabolic syndrome - “A Pilot Study.”.
283. Starnes JW, Parry TL, O’Neal SK, Bain JR, Muehlbauer MJ, Honcoop Circulation 118: 346-354, 2008. DOI: 10.1161/CIRCULATION-
A, Ilaiwy A, Christopher PM, Patterson C, Willis MS. Exercise-induced AHA.108.772822.
alterations in skeletal muscle, heart, liver, and serum metabolome iden- 303. Toledo FGS, Menshikova EV, Azuma K, Radiková Z, Kelley CA, Ritov
tified by non-targeted metabolomics analysis. Metabolites 7: 40, 2017. VB, Kelley DE. Mitochondrial capacity in skeletal muscle is not stim-
ulated by weight loss despite increases in insulin action and decreases
DOI: 10.3390/metabo7030040. in intramyocellular lipid content. Diabetes 57: 987-994, 2008. DOI:
284. Stewart KJ. Exercise training and the cardiovascular consequences 10.2337/db07-1429.
of type 2 diabetes and hypertension: Plausible mechanisms for 304. Toledo FGS, Menshikova EV, Ritov VB, Azuma K, Radikova Z,
improving cardiovascular health. JAMA 288: 1622-1631, 2002. DOI: DeLany J, Kelley DE. Effects of physical activity and weight loss
10.1001/jama.288.13.1622. on skeletal muscle mitochondria and relationship with glucose
285. Stewart LK, Flynn MG, Campbell WW, Craig BA, Robinson JP, control in type 2 diabetes. Diabetes 56: 2142-2147, 2007. DOI:
McFarlin BK, Timmerman KL, Coen PM, Felker J, Talbert E. Influ- 10.2337/db07-0141.
ence of exercise training and age on CD14+ cell-surface expression 305. Tomas-Carus P, Ortega-Alonso A, Pietilainen KH, Santos V,
of toll-like receptor 2 and 4. Brain Behav Immun 19: 389-397, 2005. Goncalves H, Ramos J, Raimundo A. A randomized controlled trial on
DOI: 10.1016/j.bbi.2005.04.003. the effects of combined aerobic-resistance exercise on muscle strength
286. Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO, and fatigue, glycemic control and health-related quality of life of
Gonzalez M, Yancopoulos GD, Glass DJ. The IGF-1/PI3K/Akt path- type 2 diabetes patients. J Sports Med Phys Fitness 56: 572-578, 2016.
way prevents expression of muscle atrophy-induced ubiquitin ligases 306. Tufescu A, Kanazawa M, Ishida A, Lu H, Sasaki Y, Ootaka T,
by inhibiting FOXO transcription factors. Mol Cell 14: 395-403, 2004. Sato T, Kohzuki M. Combination of exercise and losartan enhances
DOI: 10.1016/S1097-2765(04)00211-4. renoprotective and peripheral effects in spontaneously type 2 diabetes
287. Stolk RP, Vingerling JR, de Jong PT, Dielemans I, Hofman A, Lam- mellitus rats with nephropathy. J Hypertens 26: 312-321, 2008. DOI:
berts SW, Pols HA, Grobbee DE. Retinopathy, glucose, and insulin in 10.1097/HJH.0b013e3282f2450b.
an elderly population: The Rotterdam Study. Diabetes 44: 11-15, 1995. 307. Turcotte LP, Fisher JS. Skeletal muscle insulin resistance: Roles of fatty
DOI: 10.2337/diab.44.1.11. acid metabolism and exercise. Phys Ther 88: 1279-1296, 2008. DOI:
288. Streuber SD, Amsterdam EA, Stebbins CL. Heart rate recovery in heart 10.2522/ptj.20080018.
failure patients after a 12-week cardiac rehabilitation program. Am J 308. Turcotte LP, Richter EA, Kiens B. Increased plasma FFA uptake
Cardiol 97: 694-698, 2006. DOI: 10.1016/j.amjcard.2005.09.117. and oxidation during prolonged exercise in trained vs. untrained
289. Swain RA, Harris AB, Wiener EC, Dutka MV, Morris HD, Theien humans. Am J Physiol Endocrinol Metab 262: E791-E799, 1992. DOI:
BE, Konda S, Engberg K, Lauterbur PC, Greenough WT. Prolonged 10.1152/ajpendo.1992.262.6.E791.
exercise induces angiogenesis and increases cerebral blood volume in 309. U. K. Prospective Diabetes Study Group. U.K. Prospective Diabetes
primary motor cortex of the rat. Neuroscience 117: 1037-1046, 2003. Study 16: Overview of 6 years’ therapy of Type II Diabetes: A pro-
DOI: 10.1016/S0306-4522(02)00664-4. gressive disease. Diabetes 44: 1249-1258, 1995. DOI: 10.2337/diab.
290. Sylow L, Tokarz VL, Richter EA, Klip A. The many actions of insulin 44.11.1249.
in skeletal muscle, the paramount tissue determining glycemia. Cell 310. Umpierre D, Ribeiro PAB, Kramer CK, Leitão CB, Zucatti ATN,
Metab 33: 758-780, 2021. DOI: 10.1016/j.cmet.2021.03.020. Azevedo MJ, Gross JL, Ribeiro JP, Schaan BD. Physical activity
291. Szczerbinski L, Golonko A, Taylor M, Puchta U, Konopka P, Paszko A, advice only or structured exercise training and association With HbA1c
Citko A, Szczerbinski K, Gorska M, Zabielski P, Błachnio-Zabielska levels in type 2 diabetes: A systematic review and meta-analysis. JAMA
A, Larsen S, Kretowski A. Metabolomic profile of skeletal muscle 305: 1790-1799, 2011. DOI: 10.1001/jama.2011.576.
and its change under a mixed-mode exercise intervention in progres- 311. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection
sively dysglycemic subjects. Front Endocrinol 12: 778442, 2021. DOI: from obesity-induced insulin resistance in mice lacking TNF-α func-
10.3389/fendo.2021.778442. tion. Nature 389: 610-614, 1997. DOI: 10.1038/39335.
292. Szczerbinski L, Taylor MA, Puchta U, Konopka P, Paszko A, Citko 312. Vaynman S, Ying Z, Gomez-Pinilla F. Hippocampal BDNF mediates
A, Szczerbinski K, Goscik J, Gorska M, Larsen S, Kretowski A. The the efficacy of exercise on synaptic plasticity and cognition. Eur J Neu-
response of mitochondrial respiration and quantity in skeletal muscle rosci 20: 2580-2590, 2004. DOI: 10.1111/j.1460-9568.2004.03720.x.
and adipose tissue to exercise in humans with prediabetes. Cell 10: 313. Vendrell J, El Bekay R, Peral B, García-Fuentes E, Megia A, Macias-
3013, 2021. DOI: 10.3390/cells10113013. Gonzalez M, Real JF, Jimenez-Gomez Y, Escoté X, Pachón G,
Simó R, Selva DM, Malagón MM, Tinahones FJ. Study of the 325. Wilmot EG, Edwardson CL, Achana FA, Davies MJ, Gorely T, Gray
potential association of adipose tissue GLP-1 receptor with obesity LJ, Khunti K, Yates T, Biddle SJH. Sedentary time in adults and the
and insulin resistance. Endocrinology 152: 4072-4079, 2011. DOI: association with diabetes, cardiovascular disease and death: System-
10.1210/en.2011-1070. atic review and meta-analysis. Diabetologia 55: 2895-2905, 2012. DOI:
314. Veugen MGJ, Linssen PBC, Henry RMA, Koster A, Kroon AA, 10.1007/s00125-012-2677-z.
Stehouwer CDA, Brunner-La Rocca H. Measures of left ventricular 326. Wing RR, Lang W, Wadden TA, Safford M, Knowler WC, Bertoni
diastolic function and cardiorespiratory fitness according to glucose AG, Hill JO, Brancati FL, Peters A, Wagenknecht L, the Look
metabolism status: The Maastricht Study. J Am Heart Assoc Car- AHEAD Research Group. Benefits of modest weight loss in improving
diovasc Cerebrovasc Dis 10: e020387, 2021. DOI: 10.1161/JAHA. cardiovascular risk factors in overweight and obese individuals
120.020387. with type 2 diabetes. Diabetes Care 34: 1481-1486, 2011. DOI:
315. Vinik AI, Ziegler D. Diabetic cardiovascular autonomic neuropathy. 10.2337/dc10-2415.
Circulation 115: 387-397, 2007. DOI: 10.1161/CIRCULATION- 327. Winn NC, Cottam MA, Wasserman DH, Hasty AH. Exercise and adi-
AHA.106.634949. pose tissue immunity: Outrunning inflammation. Obesity 29: 790-801,
316. Vogelzangs N, van der Kallen CJH, van Greevenbroek MMJ, van der 2021. DOI: 10.1002/oby.23147.
Kolk BW, Jocken JWE, Goossens GH, Schaper NC, Henry RMA, 328. Wolff G, Esser KA. Scheduled exercise phase shifts the circadian clock
Eussen SJPM, Valsesia A, Hankemeier T, Astrup A, Saris WHM, Ste- in skeletal muscle. Med Sci Sports Exerc 44: 1663-1670, 2012. DOI:
houwer CDA, Blaak EE, Arts ICW, Diogenes consortium. Metabolic 10.1249/MSS.0b013e318255cf4c.
profiling of tissue-specific insulin resistance in human obesity: Results 329. Xing W, Yan W, Fu F, Jin Y, Ji L, Liu W, Wang L, Lv A, Duan Y,
from the Diogenes study and the Maastricht Study. Int J Obes 2005 Zhang J, Zhang H, Gao F. Insulin inhibits myocardial ischemia-induced
(44): 1376-1386, 2020. DOI: 10.1038/s41366-020-0565-z. apoptosis and alleviates chronic adverse changes in post-ischemic
317. Volaco A, Cavalcanti AM, Filho RP, Précoma DB. Socioeco- cardiac structure and function. Apoptosis 14: 1050-1060, 2009. DOI:
nomic status: The missing link between obesity and diabetes 10.1007/s10495-009-0378-y.
mellitus? Curr Diabetes Rev 14: 321-326, 2018. DOI: 10.2174/ 330. Yang L, Li D-X, Cao B-Q, Liu S-J, Xu D-H, Zhu X-Y, Liu Y-J. Exercise
1573399813666170621123227. training ameliorates early diabetic kidney injury by regulating the H2
318. Walsh NP, Gleeson M, Shephard RJ, Gleeson M, Woods JA, Bishop S/SIRT1/p53 pathway. FASEB J Off Publ Fed Am Soc Exp Biol 35:
NC, Fleshner M, Green C, Pedersen BK, Hoffman-Goetz L, Rogers CJ, e21823, 2021. DOI: 10.1096/fj.202100219R.
Northoff H, Abbasi A, Simon P. Position statement. Part one: Immune 331. Yang P, Swardfager W, Fernandes D, Laredo S, Tomlinson G, Oh
function and exercise. Exerc Immunol Rev 17: 6-63, 2011. PI, Thomas S. Finding the optimal volume and intensity of resis-
319. Wang S, Chen L, Zhang L, Huang C, Xiu Y, Wang F, Zhou C, Luo tance training exercise for type 2 diabetes: The FORTE study, a
Y, Xiao Q, Tang Y. Effects of long-term exercise on spatial learning, randomized trial. Diabetes Res Clin Pract 130: 98-107, 2017. DOI:
memory ability, and cortical capillaries in aged rats. Med Sci Monit 10.1016/j.diabres.2017.05.019.
Int Med J Exp Clin Res 21: 945-954, 2015. DOI: 10.12659/MSM. 332. Yates T, Edwardson CL, Henson J, Gray LJ, Ashra NB, Troughton J,
893935. Khunti K, Davies MJ. Walking away from type 2 diabetes: A clus-
320. Ward KM, Mahan JD, Sherman WM. Aerobic training and diabetic ter randomized controlled trial. Diabet Med 34: 698-707, 2017. DOI:
nephropathy in the obese Zucker rat. Ann Clin Lab Sci 24: 266-277, 10.1111/dme.13254.
1994. 333. Yates T, Griffin S, Bodicoat DH, Brierly G, Dallosso H, Davies MJ,
321. Watson EL, Greening NJ, Viana JL, Aulakh J, Bodicoat DH, Barratt Eborall H, Edwardson C, Gillett M, Gray L, Hardeman W, Hill S,
J, Feehally J, Smith AC. Progressive resistance exercise training in Morton K, Sutton S, Troughton J, Khunti K. PRomotion Of Physical
CKD: A feasibility study. Am J Kidney Dis 66: 249-257, 2015. DOI: activity through structured Education with differing Levels of ongoing
10.1053/j.ajkd.2014.10.019. Support for people at high risk of type 2 diabetes (PROPELS): Study
322. Way KL, Hackett DA, Baker MK, Johnson NA. The effect of regular protocol for a randomized controlled trial. Trials 16: 289, 2015. DOI:
exercise on insulin sensitivity in type 2 diabetes mellitus: A systematic 10.1186/s13063-015-0813-z.
review and meta-analysis. Diabetes Metab J 40: 253-271, 2016. DOI: 334. Zhang Q, Wei Y, Chen M, Wan Q, Chen X. Clinical analysis of risk
10.4093/dmj.2016.40.4.253. factors for severe COVID-19 patients with type 2 diabetes. J Diabetes
323. Wefers JF, Woodlief TL, Carnero EA, Helbling NL, Anthony SJ, Complicat 34: 107666, 2020. DOI: 10.1016/j.jdiacomp.2020.107666.
Dubis GS, Jakicic JM, Houmard JA, Goodpaster BH, Coen PM. 335. Zhao R-Z, Jiang S, Zhang L, Yu Z-B. Mitochondrial electron transport
Relationship between physical activity, sedentary behaviors and car- chain, ROS generation and uncoupling (Review). Int J Mol Med 44:
diometabolic risk factors during gastric bypass surgery-induced weight 3-15, 2019. DOI: 10.3892/ijmm.2019.4188.
loss. Surg Obes Relat Dis Off J Am Soc Bariatr Surg 13: 210-219, 336. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ven-
2017. DOI: 10.1016/j.soard.2016.08.493. tre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller
324. Welly RJ, Liu T-W, Zidon TM, Rowles JL, Park Y-M, Smith TN, DE. Role of AMP-activated protein kinase in mechanism of metformin
Swanson KS, Padilla J, Vieira-Potter VJ. Comparison of diet vs. action. J Clin Invest 108: 1167-1174, 2001.
exercise on metabolic function & gut micorbiota in obese rats. 337. Ziegler D. Treatment of diabetic polyneuropathy. Ann N Y Acad Sci
Med Sci Sports Exerc 48: 1688-1698, 2016. DOI: 10.1249/MSS. 1084: 250-266, 2006. DOI: 10.1196/annals.1372.008.
0000000000000964.