1 Hepatic AMPK signaling activation in response to dynamic REDOX balance is a 2 biomarker of exercise to improve blood glucose control 3 4 Meiling Wu†1, Anda Zhao†1, Xingchen Yan†1, Hongyang Gao†2, Chunwang Zhang1, 5 Xiaomin Liu1, Qiwen Luo1, Feizhou Xie3, Shanlin Liu‡1,4, Dongyun Shi‡1 6 7 Affiliations 8 1 9 Fudan University, Shanghai, 200032, People's Republic of China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, 10 2 11 Shanghai, 200032, People's Republic of China 12 3 13 Shanghai, 200032, People's Republic of China 14 4 15 200032, People's Republic of China Institute of Electronmicroscopy, School of Basic Medical Sciences, Fudan University, Changning Maternity and Infant Health Hospital, East China Normal University, Free Radical Regulation and Application Research Center of Fudan University, Shanghai, 16 17 18 19 20 21 22 † Co-first authors contributed equally to this work 23 ‡ Correspondence to Prof. Dongyun Shi (Email: dyshi@fudan.edu.cn) and Prof. Shanlin 24 Liu (Email: slliu@shmu.edu.cn), Department of Biochemistry and Molecular Biology, 25 School of Basic Medical Sciences, Fudan University, NO.130 Dong’an Road, Shanghai, 26 200032, People's Republic of China, Tel: +021 54237299 Fax: +021 54237897 27 Abstract 28 29 Antioxidant intervention is considered to inhibit reactive oxygen species (ROS) and 30 alleviates hyperglycemia. Paradoxically, moderate exercise can produce ROS to improve 31 diabetes. The exact redox mechanism of these two different approaches remains largely 32 unclear. Here, by comparing exercise and antioxidants intervention on type 2 diabetic rats, 33 we found moderate exercise upregulated compensatory antioxidant capability and reached a 34 higher level of redox balance in the liver. In contrast, antioxidant intervention achieved a 35 low-level redox balance by inhibiting oxidative stress. Both of these two interventions 36 could promote glucose catabolism and inhibit gluconeogenesis through activation of 37 hepatic AMPK signaling, therefore ameliorating diabetes. During exercise, different levels 38 of ROS generated by exercise have differential regulations on the activity and expression of 39 hepatic 40 glutathionylation activation. However, excessive exercise increased oxidative damage and 41 inhibited the activity and expression of AMPK. Overall, our results illustrate that both 42 exercise and antioxidant intervention improve blood glucose in diabetes by promoting 43 redox balance, despite different levels of redox balance. These results indicate that the 44 AMPK signaling activation, combined with oxidative damage markers, could act as a 45 sensitive biomarker, reflecting the threshold of redox balance defining effective treatment 46 in diabetes. These findings provide theoretical evidence for the precise treatment of 47 diabetes by antioxidants and exercise. AMPK. Moderate exercise-derived ROS promoted hepatic 48 49 50 Key words 51 Redox balance; AMPK signaling; exercise; antioxidant; glutathionylation; diabetes 52 AMPK 53 Introduction 54 55 Diabetes mellitus is a chronic metabolic disorder disease, which has emerged as a global 56 public health problem. According to the latest epidemiological data from the International 57 Diabetes Federation, the global diabetes prevalence in 20–79 year olds was estimated to be 58 10.5% (536.6 million people) in 2021, and is expected to rise to 12.2% (783.2 million) in 59 2045 (1). With the development of genomics, proteomics and metabolomics, it has been 60 discovered by many studies that type 2 diabetes is associated with irreversible risk factors 61 such as age, genetics, race, and ethnicity and reversible factors such as diet, physical 62 activity and lifestyle (2, 3). 63 64 Aerobic metabolism in glucose oxidation, mitochondrial damage and oxidative stress have 65 been considered to play a critical role in the occurrence and development of diabetes (4). 66 Exercise and antioxidant supplements are often suggested as essential therapeutic strategies 67 in the early stages of type 2 diabetes (5, 6), with different mechanisms. It has been reported 68 that chronic exercise training can alleviate oxidative stress and diabetic symptoms by 69 improving cellular mitochondrial function and biogenesis in the diabetic state (7). 70 Contradictorily, exercise also increases ROS production, while prolonged or high-intensity 71 exercise could result in mitochondrial functional impairment to aggravate complications of 72 diabetes (8). Since 1970s, studies have demonstrated that 1 hour of moderate endurance 73 exercise can increase lipid peroxidation in humans (9, 10). In 1998, Ashton directly 74 detected increasing free radical levels in exercising humans using electron paramagnetic 75 resonance spectroscopy (EPR) and spin capture (11). These results led to a great deal of 76 interest in the role of ROS in physical exercise (12-14). Regarding the contradiction of 77 exercise on ROS scavenging or production, James D Watson also hypothesized that type 2 78 diabetes is accelerated by insufficient oxidative stress rather than oxidative stress (15), 79 based on the effect of exercise on diabetes treatment. Although Watson’s opinions 80 supported that exercise could treat diabetes by producing ROS, whether exercise-induced 81 ROS production is beneficial or detrimental to diabetes is still being debated. The specific 82 regulation of ROS produced by exercise on diabetic blood glucose in vivo is unclear. In 83 contrast, the general view of the antioxidant treatment for diabetes is that antioxidants 84 reduce cytotoxic ROS and oxidation products, thus alleviating diabetes and achieving 85 glycemic control (16). Our previous study also found that hepatic mitochondrial ROS 86 scavengers and antioxidant substances inhibited the oxidative products such as MDA and 4- 87 HNE in diabetic animals and favored glycemic control (17, 18). Exercise-induced oxidation 88 and antioxidant administration, as two opposite approaches, could achieve the regulation of 89 diabetes, respectively. However, the differences in redox mechanisms between these two 90 approaches to diabetes treatment have not been fully understood. 91 92 It is well established that the increase of skeletal muscle glucose uptake during exercise is 93 crucial in glycemic control (19-21). Considering that liver is another vital organ for 94 maintaining blood glucose homeostasis, including storing, utilizing and producing glucose, 95 exercise-induced hepatic redox metabolism is also significant. The activation of hepatic 96 AMP-activated protein kinase (AMPK), which acts as a ‘metabolic master switch’, 97 alleviates diabetes symptoms by reducing glycogen synthesis, increasing glycolysis, and 98 promoting glucose absorption in surrounding tissues (22). Therefore, the activation of 99 AMPK in the liver is significant for regulating glucose and lipid metabolism in the blood. 100 Zmijewski et al. found that AMPK could be activated by hydrogen peroxide stimulation 101 through direct oxidative modification (23). In contrast, other studies suggested that 102 oxidative stress could disrupt the activation of the AMPK signaling pathway (24, 25). Our 103 previous study explored the mechanism by which redox status contributes to hepatic 104 AMPK dynamic activation. Under a low ROS microenvironment, GRXs mediated S- 105 glutathione modification activates AMPK to improve glucose utilization. In contrast, under 106 an excessive ROS microenvironment, sustained high level ROS might cause loss of AMPK 107 protein (26). These studies indicate that oxidative modification can directly regulate AMPK 108 activity in liver cells, thus activating downstream signaling pathways to regulate glucose 109 and lipid metabolism. However, it is unclear why two seemingly contradictory 110 phenomenon of antioxidant intervention and exercise-induce ROS can promote AMPK 111 activation. Moderate exercise has been proved significantly elevate systemic oxidative 112 stress. At the same time, endogenous antioxidant defences also increased to counteract 113 increased levels of ROS induced by exercise (27). Thus, we hypothesized that both 114 antioxidants and exercise could reach either high-level or low-level redox balance in 115 diabetic individuals. Moreover, the activity and expression of AMPK might be a marker of 116 redox balance in vivo (Fig. 1). 117 118 Hence, the present study was designed to understand the different mechanisms of exercise 119 and antioxidant intervention in diabetes and verify the activation of hepatic AMPK as a 120 hallmark of dynamic redox balance. Firstly, we utilized the streptozotocin-high fat diet 121 (STZ-HFD) induced type 2 diabetic (T2DM) model in rats to clarify the hepatic redox 122 status in T2DM rats after the exercise or antioxidant intervention. Then, according to the 123 exercise intensity and mode, we divided the exercise groups into three modes and found 124 that AMPK activation could serve as a biomarker of redox balance and moderate exercise 125 in diabetic treatment. In this study, we found that AMPK activation and its downstream 126 pathways could reflect the threshold of exercise or antioxidant administration for diabetes 127 treatment. This study provides clues for the personalized treatment of diabetes by 128 antioxidants and exercise. 129 130 Results 131 132 1. Exercise promotes antioxidant levels through producing ROS, leading to a high 133 level of REDOX balance in the liver. 134 135 To investigate the hepatic redox regulation in diabetes after exercise intervention, we 136 established the T2DM rat model by feeding HFD followed by a low dose of STZ injection 137 (35 mg/kg). The exercise intervention was started from Day 0 to Day 28 (Fig. 2A). 138 According to previous studies, the initial speed of exercise was 15 m/min, and the speed 139 was increased by 3 m/min every 5 min. After the speed reached 20 m/min, the speed was 140 maintained for another 60 min with slope of 5%. The exercise intensity was 64%-76% 141 VO2max (28). The low-intensity continuous exercise (CE) can be regarded as aerobic 142 exercise. 143 The ROS-generating NADPH oxidases (NOXs) have been recognized as one of the main 144 sources of ROS production in cells (29). Cyclooxygenase 2 (COX2) activity could also act 145 as a stimulus for ROS production (30). The expressions of NOX4 and COX2 in the liver 146 were increased in the diabetic group. After exercise intervention, NOX4 and COX2 level 147 were further up-regulated compared with the diabetic group (Fig. 2B-D). These results 148 indicate the exercise intervention up-regulated ROS production. 149 Next, we detected the expression of antioxidant enzymes in liver tissue. Nrf2 is the central 150 regulator of the threshold mechanisms of oxidative stress and ROS generation (31). With 151 the increase of ROS level in the development of diabetes, Nrf2 was activated to induce the 152 transcription of several antioxidant enzymes (32, 33). We found an increase in Nrf2 153 expression in diabetic rats (Fig. 2E-F). After CE intervention, the level of Nrf2 levels 154 further increased, indicating that exercise intervention could activate antioxidant system 155 (Fig. 2E-F). Under stress conditions, Nrf2 translocates to the nucleus and binds to 156 antioxidant response elements (AREs), which results in the expression of diverse 157 antioxidant and metabolic genes, such as thioredoxin (Trx), to relieve oxidative damage 158 (34). Thioredoxin-1 (Trx-1), a type of cytosolic isoform of Trx, has been widely accepted 159 to regulate glutathione metabolism with GRX and PRX. After CE intervention, we found 160 the protein expression of GRX1 and TRX1 were up-regulated (Fig. 2H, 2J-K). Notably, the 161 PRX expression also showed a trend of increase (Fig. 2H-1I). Sestrin2 is a cysteine sulfinyl 162 reductase that plays crucial roles in regulation of antioxidant actions (35). As an 163 endogenous antioxidant, the hepatic sestrin2 level also showed a significant increase in the 164 CE group (Fig. 2E,2G). 165 Since excess ROS can cause the increase of oxidative damage (36), we further detected the 166 protein damage and lipid peroxidation to determine the redox status. 3-Nitrotyrosine (3-NT) 167 and protein carbonylation are biomarkers of reactive nitrogen species (RNS) and ROS 168 modified proteins (37, 38). We found that the CE intervention reduced the 3-Nitrotyrosine 169 level and did not further decrease the protein carbonylation level (Fig. 2L-N). MDA, a 170 biomarker of lipid peroxidation, was also significantly up-regulated in the diabetic group 171 but decreased in exercise group (Fig. 2O). These results indicate that the high ROS 172 production in the CE group could, instead, increase the antioxidant status to avoid oxidative 173 damage. It suggests that CE can promote redox to reach a high level of balance. Therefore, 174 even if exercise increases the ROS-generating enzymes NOX4 and COX2, the increase in 175 ROS production does not lead to oxidative damage. 176 177 2. Antioxidant intervention alleviates blood glucose through reducing oxidative stress, 178 leading to a low level of REDOX balance in the liver. 179 Recent studies have suggested that NADPH oxidase is one of the primary sources of ROS 180 (29, 39). Apocynin has already been characterized as an NADPH oxidase inhibitor in the 181 early 1980s, and it can also act as an antioxidant (40). Our previous study showed that 182 apocynin intervention alleviated blood glucose by inhibiting oxidative products. In this 183 study, the antioxidant supplement was also started from Day 0 to Day 28 in this study (Fig. 184 3A). We found that apocynin supplement decreased the protein carbonylation level and 185 MDA level in the liver (Fig. 3B-C). Also, the TAOC level was increased after apocynin 186 treatment, indicating the decrease of oxidation level (Fig. 3D). Moreover, as endogenous 187 antioxidant, the Sestrin2 and Nrf2 expression decreased after apocynin intervention (Fig. 188 3E-G). These results indicate that the antioxidant intervention reduced the ROS in diabetic 189 hepatocytes, thereby decreasing the ROS-induced compensatory upregulation of Sestrin2 190 and Nrf2. 191 Consistently, Glut2, a glucose sensor in the liver, was increased in diabetic liver and 192 decreased after the apocynin supplement (Fig. 3E and 3H). The postprandial blood glucose 193 , fasting blood glucose and oral glucose tolerance (2 h after oral glucose, OGTT) were 194 decreased in the apocynin intervention group compared with the diabetic rat group (Fig. 3I- 195 K). Consistent with the apocynin intervention group, the exercise group also showed lower 196 postprandial blood glucose and fasting blood glucose levels and OGTT (Fig. 3I-K). These 197 studies indicate that the apocynin treatment improved the diabetes through inhibiting ROS 198 level and protein oxidative damage to achieve a low-level redox balance. 199 200 3. Moderate exercise-generated ROS promotes activation of AMPK by 201 phosphorylation and reduces blood glucose level, while excessive exercise- generated 202 oxidative stress reduces AMPK expression and exacerbates diabetes. 203 In order to find out the biomarkers that could reflect moderate exercise to improve blood 204 glucose control, diabetic rats were divided into short-term continuous exercise (CE), 205 intermittent exercise (IE), and excessive exercise (EE) according to the exercise intensity 206 and mode (28). We found that the random blood glucose and 2h OGTT in CE and IE 207 treated diabetic rats were decreased (Fig. 4A-B). In contrast, EE intervention did not 208 improve blood glucose but slightly increased random and 2h OGTT (Fig. 4A-B). 209 We detected the increase of ROS production-related enzymes, such as NOX4 and COX2 in 210 the EE group, indicating the highest oxidation level (Fig. 4C-E). Next, we detected the 211 expression of antioxidant enzymes and oxidative damage in the liver tissue of exercise- 212 treated T2D rats. As shown in Fig. 4F-G, IE intervention increased the activity of MnSOD 213 as shown by decreased level of acetylation compared with the diabetic rats. The expression 214 of GRX and TRX were up-regulated after CE intervention (Fig. 4F, 4H-I). Furthermore, we 215 detected the oxidative damage in these three modes. The results showed that the CE and IE 216 group did not obviously change the protein carbonylation level. However, the EE 217 intervention promoted the protein carbonylation in the liver, indicating this mode of action 218 is not due to free radical scavenging but oxidative damage (Fig. 4J). In addition to the 219 protein damage, hepatic MDA concentration showed significant up-regulation in the 220 diabetic group but decreased in CE and IE group (Fig. 4K), while increased MDA in the EE 221 group indicates oxidative damage. Among these three exercise modes, the IE group showed 222 the lowest level of oxidation (the minor increase in NOX4 and a slight decrease in 223 carbonylation). Although the levels of antioxidant enzymes such as GRX and TRX did not 224 increase, the activity of MnSOD also increased significantly (Fig. 4F-4G). The reduction of 225 MDA level also indicates IE group did not form oxidative damage (Fig. 4K), indicating the 226 IE group could also maintain a relatively high level of redox balance. Nevertheless, the 227 decrease of antioxidant enzymes and increase of oxidative damage in the EE group 228 indicates that the REDOX balance in the EE group was disrupted. 229 Notably, the phosphorylation of AMPK showed different patterns in three kinds of 230 exercise, among which both CE and IE intervention could promote the phosphorylation of 231 AMPK compared to the diabetic rats (Fig. 4M-N). EE intervention did not increase the 232 content of AMPK phosphorylation, which might be caused by the reduction of AMPK 233 level. Meanwhile, the ratio of AMP to ATP was detected, and exercise-activated AMPK 234 did not exhibit AMP-dependent characteristics at this time (Fig. 4L). These results suggest 235 that moderate exercise-generated ROS may directly promote AMPK activation by 236 phosphorylation without AMP upregulation and reduce blood and liver glucose levels. 237 However, excessive exercise-generated oxidative stress reduces AMPK expression and 238 exacerbates diabetes. 239 4. Moderate exercise promoted glycolysis and mitochondrial tricarboxylic acid cycle 240 and inhibited the gluconeogenesis in the liver of diabetic rats. 241 Next, we further explored the mechanism by which inhibiting blood glucose during CE and 242 IE intervention. Fructose-2,6-diphosphate (F-2,6-P2; also known as F-2,6-BP), which is a 243 product of the bifunctional enzyme 6-phosphofructose 2-kinase/fructose 2,6-diphosphatase 244 2 (PFK/FBPase 2, also known as PFKFB2), is a potent regulator of glycolytic and 245 gluconeogenic flux. The phospho-PFKFB2 to PFKFB2 ratio represents the glycolytic rate. 246 A high ratio of phospho-PFKFB2:PFKFB2 leads to an increase in the F-2,6-P2 level and 247 the allosteric activation of phosphor-fructose kinase 1 (PFK1), while a low ratio leads to a 248 decrease in F-2,6-P2 and an increase in gluconeogenesis (41). The overexpression of 249 bifunctional enzymes in mouse liver can reduce blood glucose levels by inhibiting hepatic 250 glucose production (42). Therefore, bifunctional enzymes are also a potential target for 251 reducing hepatic glucose production. In our study, the p-PFK2:PFK2 ratio decreased in the 252 diabetic rats but was enhanced by CE and IE intervention (Fig. 5A-C), suggesting that CE 253 and IE could reverse gluconeogenesis to glycolysis by enhancing PFK/FBPase. Meanwhile, 254 the substrates of the glycolytic pathway (such as DHAP, Fig. 5D) and the tricarboxylic acid 255 cycle (such as citrate, succinate and malate, Fig. 5D) showed an upward trend. 256 Besides glycolysis, gluconeogenesis are critical in maintaining liver and blood glucose 257 homeostasis. FoxO1 has been tightly linked with hepatic gluconeogenesis through 258 inhibiting the transcription of gluconeogenesis-related PEPCK and G6Pase expression (43, 259 44). Herein, we found the expression of FoxO1 was increased in the diabetic group but 260 reduced in the CE and IE groups (Fig. 5E-F). Meanwhile, the mRNA level of Pepck and 261 G6PC also decreased in the CE and IE groups (Fig. 5H-I). These results indicate that 262 moderate exercise (CE and IE) inhibited gluconeogenesis through down-regulating FoxO1. 263 For the glucose uptake, we detected the protein expression of GLUT2 in the liver tissue, 264 which helps in the uptake of glucose by the hepatocytes for glycolysis and glycogenesis. 265 We found the level of GLUT2 was increased in diabetic mice, but down-regulated by the 266 CE and IE intervention (Fig. 5E and 5G). Taken together, these results illustrated that 267 moderate exercise promoted glucose transport, glucose catabolism and inhibited the 268 gluconeogenesis in the liver of diabetic rats (Fig. 5J). 269 270 5. Moderate exercise inhibited hepatic mitophagy, while excessive exercise exhibited 271 opposite effect and inhibited the mitochondrial biogenesis. 272 The electron transport associated with the mitochondrial function is considered the major 273 process leading to ROS production during exercise (45). To further explore the downstream 274 signal of AMPK activation in moderate and excessive exercise, we detected the protein 275 expression of mitochondrial dynamic and mitochondrial biogenesis. According to the result 276 in Fig. 6A-E, we found that the mitochondrial fusion protein MFN was significantly 277 decreased in the liver of the excessive exercise group, and the mitochondrial fission protein 278 (Fis) and autophagy-related protein ATG5 and LC3B did not change, compared with the 279 diabetic group. Notably, the ATG5 and LC3B levels decreased in the CE and IE group, 280 compared with the diabetic group (Fig. 6A-E). Since PGC-1α is a transcriptional 281 coactivator, a central inducer of mitochondrial biogenesis in cells (46), we measured the 282 expression of PGC-1α and found that its expression was increased in CE and IE group, but 283 not in the EE group (Fig. 6F). 284 These results indicates that moderate exercise promoted mitochondrial biogenesis and 285 ameliorated autophagy in the liver. However, excessive exercise aggravated mitochondrial 286 fission and did not alleviate autophagy. Similarly, the mitochondria structure of the live 287 tissue in the EE group was fragmented and showed greatly diminished cristae and swelling 288 matrix under transmission electron microscopy, reflecting a defect in oxidative 289 phosphorylation. However, the CE and IE group showed increased numbers of cristae and a 290 clear structure of mitochondrial cristae (Fig. 6H). These results all showed that the in vivo 291 mitochondrial ROS burst caused by excessive exercise inhibited the expression of AMPK 292 and promoted mitophagy. The damage to the mitochondrial dynamics and structure in liver 293 tissue led to abnormal aerobic oxidation, thereby aggravating diabetes. 294 6. Moderate ROS activates AMPK through GRX-mediated glutathionylation 295 To further illustrate the effect of moderate ROS on the activation of AMPK, primary 296 hepatocytes were intervened with H2O2 to mimic ROS production in vivo. H2DCFDA and 297 dihydroethidium dyes were used to assess intracellular ROS levels in the H2O2-treated 298 primary hepatocytes. As shown in Fig. 7A-7B, the levels of ROS (DCF), such as H2O2, and 299 O2•− accumulation (dihydroethidium) were dose-dependently increased in the primary 300 hepatocytes treated with 50-200 μM H2O2. Consistent with our previous study (26), we 301 found moderate ROS would activate AMPK through GRX-mediated S-glutathionylation. 302 As shown in Fig. 7C, exposure to 50 and 100 μmol/l H2O2 led to an increase of GSS- 303 protein adduct, concomitant with the AMPK phosphorylation (Fig. 7C-F), suggesting that 304 the ROS level within redox balance threshold could induce glutathionylation and 305 phosphorylation of AMPK to activate AMPK. However, when the concentration of ROS 306 was higher (200 μmol/l H2O2), both of AMPK glutathionylation and phosphorylation were 307 decreased (Fig. 7E-7H). These results indicates that activation of AMPK by moderate ROS 308 might be mediated through GRX-mediated S-glutathionylation. 309 Meanwhile, we also detected the substrates of glycolysis and aerobic oxidation at different 310 concentrations of H2O2. We found the exposure to 20-100 μmol/l H2O2, which made cells 311 within the redox balance threshold, showed a trend of increase on glycolysis and aerobic 312 oxidation substrates, indicating the increase of hepatic glucose catabolism (data not shown). 313 314 Discussion 315 316 It has been shown that both antioxidants and exercise can be beneficial in substantially 317 ameliorating hyperglycaemia through ROS-mediated mechanisms in diabetes patients. 318 However, antioxidant intervention reduces oxidative stress, while exercise produces ROS. 319 It is imperative to explore the mechanisms underlying these seemingly paradoxical 320 approaches for effective diabetes treatment. 321 The remission of diabetes by antioxidant intervention has been well documented. Some 322 compounds in food that have substantial antioxidant activities or inhibit NADPH oxidase, 323 such as polyphenols and flavonoids (47), have been shown to improve blood glucose and 324 relieve type 2 diabetes in animal experiments. Several clinical trials also demonstrated the 325 relief of diabetic hyperglycaemia by antioxidants (48, 49). Our previous study found that 326 hepatic mitochondrial ROS scavenger and antioxidant substances inhibited the oxidative 327 products such as MDA and 4-HNE in diabetic mice and rats and improved blood glucose 328 control (18). These results indicate that reducing the oxidative level of diabetic animals 329 could treat diabetes. Hepatic AMPK regulates cellular and whole-body energy homeostasis, 330 signals to stimulate glucose uptake in skeletal muscles, fatty acid oxidation in adipose and 331 other tissues, and reduces hepatic glucose production (50, 51). Numerous pharmacological 332 agents, including the first-line oral drug metformin, natural compounds, and hormones are 333 known to activate AMPK (52-55). Moreover, our previous study found that antioxidant 334 intervention in diabetic rats could promote the phosphorylation and activation of AMPK 335 protein, thereby regulating hepatic glucose metabolism (56). Taken together, we found that 336 the activation of AMPK by antioxidant intervention was accompanied by a decrease in 337 oxidative stress level in diabetic rats, resulting in a low level of redox balance to benefit 338 diabetic hyperglycaemia. 339 In the meaning time, regular exercise have been recommended to mitigate symptoms of 340 many diseases, including psychiatric, neurological, metabolic, cardiovascular, pulmonary, 341 musculoskeletal, and even cancer (57). John Holloszy’s studies found that exercise 342 improved insulin sensitivity in patients with type 2 diabetes and provided a better 343 understanding of how muscle adapts to endurance exercise (19-21, 58-60). Although the 344 benefits of exercise are irrefutable, excessive exercise is harmful (8), suggesting the 345 importance of the amount and intensity of exercise. Recently, Chrysovalantou et al. found 346 that NADPH oxidase 4 (NOX4) is a crucial exercise-related protein to regulate adaptive 347 responses and prevent insulin resistance (61). We found that exercise could indeed increase 348 NOX4 expression, but NOX4 was also upregulated in excessive exercise. Although 349 Chrysovalantou's study highlights the role of the redox environment in exercise, the 350 biomarkers of effective intervention of moderate exercise on diabetes remain unclear. 351 352 The mechanisms of exercise for the treatment of diabetes have previously been studied 353 mainly around the skeletal muscle, as the activation of AMPK in skeletal muscle during 354 exercise is considered mainly caused by the increase of intracellular AMP:ATP ratio and 355 phosphorylation of Thr172 on the “activation loop”7 of the α-subunit (62). The activation 356 of AMPK leads to the inhibition of mTORC1 activity and activation of PGC-1α, which 357 enhances mitochondrial biogenesis and further increases muscle uptake of glucose from the 358 blood (63). However, liver energy state also plays an essential role in the activation of 359 AMPK (64) and liver is known to be critical in whole-body glucose tolerance (65). In 360 addition, AMPK is also considered as a redox-sensitive protein, and its cysteine 299 and 361 304 sites are likely to be regulated by the oxidation of hydrogen peroxide (23, 66, 67). 362 Thus, AMPK might be activated not only by the increase of AMP, but also by 363 phosphorylation through ROS regulation during exercise. 364 365 Currently there is no appropriate biomarkers to differentiate moderate and excessive 366 exercises. According to the exercise intensity and mode, we divided the exercise groups 367 into three modes: CE, IE for moderate exercises and EE for excessive exercise. Our study, 368 for the first time, found that hepatic AMPK activation could act as a biomarker of dynamic 369 redox balance during exercise to improve glycaemic control in diabetic rats. ROS generated 370 by different exercise intensities could profoundly alter the cellular redox microenvironment 371 and directly regulate the activity and expression of hepatic AMPK through a redox-related 372 mechanism. Moderate exercise produced optimal ROS directly promoted AMPK activation 373 via glutathionylation in hepatocytes. The activated AMPK signaling pathway can 374 phosphorylate and activate PFK-2 to promote glycolysis and aerobic oxidation (Fig. 8A). 375 Furthermore, AMPK activation can phosphorylate and inhibit the CRCT2 and class IIa 376 HDACs pathways in the liver, thus affecting the binding of class IIa HDACs to the FOXO- 377 family of transcription factors (68, 69). Since the gluconeogenesis related mRNA 378 expression of PEPCK and G6Pase was mainly transcribed by FOXO1, the reduction of 379 FOXO transcriptional activity induced by AMPK indicates the inhibition of 380 gluconeogenesis. Therefore, under moderate exercise, the metabolic balance between 381 glycolysis and gluconeogenesis is regulated through the activation of AMPK 382 phosphorylation, which promotes glucose catabolism and inhibits gluconeogenesis. 383 However, excessive ROS inhibits the activity and expression of AMPK in hepatocytes 384 cells, which might be related with the oxidative stress induced protein degradation (Fig. 385 8A). In addition, the number of mitochondria and the function of aerobic oxidation in the 386 EE group was significantly lower than those in the moderate exercise group. In the EE 387 group, the autophagy and fission of liver mitochondria were also up-regulated as compared 388 with the moderate exercise,accompanied by the increase of MDA, an indicator of lipid 389 peroxidation damage. These results indicate that AMPK signaling and oxidative damage- 390 related index could be investigated as sensitive biomarkers for redox status changes during 391 exercise intervention in diabetic rats. This finding can be applied to analyze the thresholds 392 of redox balance that discriminates moderate and excessive exercise. 393 394 Exercise is considered to improve blood glucose by promoting ROS levels, which seems to 395 be contradictory to antioxidant interventions of inhibiting ROS. Zsolt Radak et al proposed 396 a bell-shaped dose-response curve between normal physiological function and level of ROS 397 in healthy individuals, and suggested that moderate exercise can extend or stretch the levels 398 of ROS while increases the physiological function (70). Our results validated this 399 hypothesis and further proposed that moderate exercise could produce ROS meanwhile 400 increase antioxidant enzyme activity to maintain high level redox balance according to the 401 bell-shaped curve, whereas excessive exercise would generate a higher level of ROS, 402 leading to reduced physiological function. In this study, we found the state of diabetic 403 individuals is more applicable to the description of a S-shaped curve, due to the high level 404 of oxidative stress and decreased reduction level in diabetic individuals (Fig.8B). With the 405 increase of ROS, the physiological function of diabetic individuals gradually decreases. 406 Moderate exercise shifts the S-shaped curve into a bell-shaped dose-response curve, thus 407 reducing the sensitivity to oxidative stress in diabetic individuals and restoring redox 408 homeostasis. However, with excessive exercise, ROS production increases beyond the 409 threshold range of redox balance, resulting in decreased physiological function (Fig.8B, see 410 the decreasing portion of the bell curve to the right of the apex). 411 412 Nevertheless, the antioxidant intervention increased physiological activity by reducing ROS 413 levels in diabetic individuals, restoring a bell-shaped dose-response curve at low level of 414 ROS (Fig.8B). Therefore, redox balance could be achieved either at low level of ROS 415 mediated by antioxidant intervention or at high level of ROS mediated by moderate 416 exercise, both of which were regulated by AMPK activation. Therefore, both high and low 417 levels of redox balance can lead to high physiological function as long as they are in the 418 redox balance threshold range. Then, the activation of AMPK is an important sign of 419 exercise or antioxidant intervention to obtain redox dynamic balance which helps restore 420 physiological function. Accordingly, we speculate that the antioxidant intervention based 421 on moderate exercise might offset the effect of exercise, but antioxidants could be 422 beneficial during excessive exercise. The human study also supports that supplementation 423 with antioxidants may preclude the health-promoting effects of exercise (71). Therefore, 424 personalized intervention with respect to redox balance will be crucial for the effective 425 treatment of diabetes patients. 426 427 Together, our study revealed the mechanisms underlying seemingly contradictory effects of 428 level of ROS by either antioxidant intervention or moderate exercise for the treatment of 429 diabetes. Moderate exercise promoted hepatic ROS production and up-regulated 430 antioxidant capability, achieving a high-level balance of redox state. In contrast, antioxidant 431 intervention scavenged the hepatic free radical to form a delicate low-level balance of 432 redox state. Moreover, excessive exercise led to redox imbalance due to excess ROS levels. 433 Hepatic AMPK signaling activation could act as a sign and hallmark of moderate exercise 434 and dynamic redox balance to guide appropriate exercise or antioxidant intervention. These 435 results illustrate that it is necessary to develop a moderate exercise program according to 436 the REDOX microenvironment of diabetes patients and provide theoretical evidence for the 437 precise treatment of diabetes by antioxidants and exercise. 438 439 Materials and Methods 440 441 1. Materials 442 T-AOC kit was supplied by Changzhou Redox Biological Technology Corporation 443 (Jiangsu , CN). Antibodies against Actin, Acetylated-Lysine, P-PFK2, Ace-SOD2, ATG5, 444 LC3A/B, GAPDH, MFN1 and IgG-HRP were purchased from Cell Signaling Technology 445 (USA). Antibodies against CAT, PRX1, AMPKa1, GRX1, GRX2, SOD2, HSP90, COX1, 446 COX2 and PFK2 were purchased from ProteinTech (Wuhan, CN). Antibodies against 3- 447 NT, 4HNE, NOX4 and PGC-1α were purchased from Abcam. Antibodies against p- 448 AMPKα1/α2 were purchased from SAB (Signalway Antibody, USA). The detailed 449 antibody information is shown in Supplementary file 1a. High-fat diet (HFD) were 450 purchased from Shanghai SLRC laboratory animal Company Ltd (Shanghai, China) and the 451 nutritional composition is shown in Supplementary Table S2. 452 453 2. Animal 454 Male SD rats (150–160 g body weight, 6-8 weeks) were purchased from Fudan University 455 Animal Center (Shanghai, China). Normal chow and high-fat diet (HFD) were purchased 456 from Shanghai SLRC laboratory animal Company Ltd (Shanghai, China) and the 457 nutritional composition was shown in Supplementary file 1b. All animal care and 458 experimental procedures were approved by the Fudan University Institutional Laboratory 459 Animal Ethics Committee (NO. 20170223-123). Animals were housed in a pathogen free 460 environment with 12 h dark/light cycles. 461 462 3. Establishment of diabetic rat model 463 Rats were divided into six groups in a non-blinded, randomized manner: Control (Ctl), 464 STZ+HFD diabetic rat (T2D), Continuous exercise+STZ+HFD diabetic rat (T2D+CE), 465 intermittent exercise+STZ+HFD diabetic rat (T2D+IE), excessive exercise+STZ+HFD 466 diabetic rat (T2D+EE) and Apocynin+STZ+HFD diabetic rat (T2D+APO) (n=8 per group). 467 The sample size was calculated according to the Power Curve. The diabetic rats model was 468 established by 12hr-fasting followed by intraperitoneal injection of 0.1 M streptozotocin 469 (STZ) citrate solution (pH 4.5) at a dose of 35 mg/kg for Day1, and 35 mg/kg for Day 2 at 470 the 5th week. The HFD was started from the 1st week to the 8th week. After 8 weeks of 471 intervention, the mice were sacrificed. The tissues and plasma were collected and preserved 472 at −80 °C for further analysis. 473 Rat were acclimated to treadmill running for 3 days before the initiation of the experiments 474 and the exercise training intervention was continued for 4 weeks (5 times per week). All 475 animals were randomized before the initiation of exercise tests. 476 Continuous exercise: 477 The initial speed was 15 m/min, and the speed was increased by 3 m/min every 5 min. 478 After the speed reached 20 m/min, the speed was maintained for another 60 min with slope 479 of 5%. The exercise intensity was 64%-76% VO2max (28, 72, 73). 480 Intermittent exercise: 481 The initial speed was 15 m/min, and the speed was increased by 3 m/min every 5 min. 482 After the speed reached 20 m/min, the speed was maintained for 20 min and then 5 min rest 483 at 5 m/min. The training was continued for 3 times, and the total running time is 60 min 484 with two 5 min rest with slope of 5%. 485 Excessive exercise: 486 The initial speed was 15 m/min, and the speed was increased by 3 m/min every 5 min. 487 After the speed reached 50 m/min, the speed was maintained for another 60 min with slope 488 of 5%. The exercise intensity was higher than 80% VO2max. 489 OGTT was performed in the fasting mice with intraperitoneal injection of glucose at 1 g/kg 490 of body weight, and glucose was measured at 15min, 30 min, 60 min and 120 min, 491 respectively. Blood glucose was determined by glucometer (Roche, Switzerland). 492 493 4. Cell culture 494 Normal Human Hepatic Cell Line L02 cells (Cell Bank of Chinese Academy of Sciences) 495 were grown in DMEM supplemented with 10% FBS (GIBCO, USA) in a humidified 496 incubator (Forma Scientific) at 37 °C and 5% CO2 as described previously. The medium 497 were supplemented with 10% FBS (GIBCO, USA), 2 mmol/l glutamine, 1 mmol/l sodium 498 pyruvate, 10 mmol/l HEPES, 50 μmol/l β-mercaptoethanol, 105 U/l penicillin and 499 streptomycin. Glutamine and sodium pyruvate were purchased from Sinopharm Chemical 500 Reagent Co., Ltd, HEPES were purchased from Beyotime Biotechnology (Shanghai, CN). 501 All cell lines used in the study were tested for mycoplasma and were STR profiled. 502 503 5. Flow cytometry 504 For measurement of intracellular Superoxide, primary hepatocytes were stained with 5 μM 505 hydroethidine (superoxide indicator) (Thermo Fisher Scientific, USA). Stained cells were 506 analyzed with NovoCyte Quanteon flow cytometer (Agilent Technologies, Inc.), and 507 acquired data were analyzed with NovoExpress software (Agilent Technologies, Inc.) and 508 FlowJo software (TreeStar, Ashland, OR). 509 510 6. ATP and AMP content analysis 511 Liver tissue (20–30 mg) were homogenized on ice by perchloric acid. Homogenized 512 samples were centrifuged for 12,000 rpm at 4 °C (30 min). Supernatant was then 513 neutralized with 4 M K2CO3, followed by further centrifugation for 12,000 rpm at 4 °C for 514 20 min. Supernatant was obtained for the determination of ATP and AMP content by high 515 performance liquid chromatography (HPLC). The detection wavelength was 254 nm. 516 517 7. SOD activity assay 518 SOD activity assay was carried out using a chemiluminometric detector (Lumat LB9507, 519 Berthold). Superoxide anions were generated by adding xanthine oxidase (XO) into the 520 reaction system consisting of xanthine and Lucigenin. The drop of luminescence within 521 2 min was recorded as the relative SOD activity. 522 523 8. MDA content assay 524 MDA from the oxidative polyunsaturated fatty acids (PUFA) degradation is determined by 525 the reaction of thiobarbituric acid (TBA) with MDA to generate the stable end product of 526 MDA-TBA adduct. Liver tissue lysis was reacted with TBA to form a red product which 527 can be detected using fluorometric (Ex/Em: 532/553 nm) plate reader. 528 529 9. S-Glutathionylation of AMPK and detection of GSS-AMPK adduct formation 530 GSS-AMPK adduct were measured as described previously (23, 26). Primary hepatocytes 531 (2×106 cell/well) were incubated with ethyl ester GSH-biotin (6 mM) for 1 h. Cells were 532 then washed twice with culture buffer to remove the excess of GSH and treated with H2O2 533 for 30 min. Cell lysates were prepared in the presence of N-ethylmaleimide (5 mM) and 534 then passed through Bio-Gel P10 to remove free GSH-biotin and N-ethylmaleimide. The 535 level of GSS-protein conjugates was determined using non-reducing Western blot analysis 536 with streptavidin-HRP, whereas GSS-AMPK subunit levels were measured after pull-down 537 with streptavidin-agarose (60 min at 4 °C), followed by reducing SDS-PAGE and Western 538 blot analysis with antibodies against AMPK α subunit. 539 540 10. Metabolite profiling detection 541 Cellular metabolites were extracted and analysed by LC-MS/MS. Ferulic acid was added as 542 an internal standard to metabolite extracts, and metabolite abundance was expressed 543 relative to the internal standard and normalized to cell number. Mass isotopomer 544 distribution was determined by LC-MS/MS (AB SCIEX Triple-TOF 4600) with selective 545 reaction monitoring (SRM) in positive/negative mode 546 547 11. Western blot analysis 548 Cells and tissues were lysed in a buffer containing 1% Nonidet P-40, 0.25% sodium 549 deoxycholate, 150 mmol/l NaCl, 10 mmol/l Tris, 1 mmol/l EGTA, 1% proteinase and 550 phosphatase inhibitor cocktails (Sigma-Aldrich) at 4 °C for 30 min. Cell lysates were 551 resolved by sodium dodecyl sulfate-polyacrylamide (SDS-PAGE) gel electrophoresis, 552 transferred to polyvinylidene fluoride (PVDF) membranes, and immunoblotted with 553 primary antibodies. Membranes were incubated with HRP-conjugated secondary antibodies 554 and visualized using chemiluminescent substrate (ECL; Tanon, CN) and Tanon-5200 555 Chemiluminesent Imaging System (Tanon, CN). 556 557 12. Transmission electron microscope (TEM) 558 Rat liver tissue (1 mm*1 mm) were fixed by paraformaldehyde. The samples were 559 examined with a Jeol Jem-100SV electron microscope (Japan) which was operated at 80 560 Kv after fixed by 3% glutaraldehyde in 0.1 M phosphate buffer (pH 7.3) at Institute of 561 Electron microscopy, Shanghai Medical College of Fudan University. 562 563 13. Western blot analysis 564 Cells were lysed RIPA buffer with phosphatase inhibitor at 4 °C for 30 min. Cell lysates 565 were resolved by 10% SDS-PAGE, transferred to polyvinylidene fluoride (PVDF) 566 membranes, and probed with primary antibodies. Membranes were incubated with 567 peroxidase-conjugated secondary antibodies and visualized using a chemiluminescent 568 substrate 569 Chemiluminesent Imaging System. (ECL; GE Amersham Pharmacia, Beijing, China) and Tanon-5200 570 571 14. Statistics 572 The experimental data were expressed as mean ± SEM. One-way ANOVA was used to 573 compare among groups. Data analysis was conducted by Graphpad prism 9 statistical 574 analysis software. p < 0.05 was considered statistically significant. Data are expressed as 575 means ± SEM; n = 3 for cells experiment (n = 3 represents three times of individual 576 experiment); n = 8 for animal experiment. 577 578 Acknowledgments 579 The authors thank Dr. Xiaodong Zhang from Chengdu Brilliant Pharmaceuticals for his 580 proof reading and editing of the manuscript. The authors are also indebted to Dr. Rutan 581 Zhang and Prof. Liang Qiao from Fudan University, for analysis of LC-MS/MS data. The 582 authors thank Dr. Ziqing Tang, Mr. Qi Kang, Dr. Xiaomin Liu, Mr. Yipei He, Dr. Xiao 583 Zhang, Mrs Lihan Jiang, Mr. Kelei Dong for their assistance in animal experiments. The 584 authors are also indebted to Institute of Electronmicroscopy from Fudan University, for the 585 help on electronmicroscopy analysis. 586 This work was supported by grants from the National Natural Science Foundation of China 587 (Grants No. 31770916). 588 589 Competing interests: 590 All other authors declare they have no competing interests 591 592 Data and materials availability: 593 All data are available in the main text or the supplementary materials. 594 595 References 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. H. Sun et al., IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract 183, 109119 (2022). A. H. Heald et al., Estimating life years lost to diabetes: outcomes from analysis of National Diabetes Audit and Office of National Statistics data. Cardiovasc Endocrinol Metab 9, 183-185 (2020). J. C. N. Chan et al., The Lancet Commission on diabetes: using data to transform diabetes care and patient lives. Lancet 396, 2019-2082 (2021). C. Iacobini, M. Vitale, C. Pesce, G. Pugliese, S. Menini, Diabetic Complications and Oxidative Stress: A 20-Year Voyage Back in Time and Back to the Future. Antioxidants (Basel) 10, (2021). J. P. Kirwan, J. Sacks, S. Nieuwoudt, The essential role of exercise in the management of type 2 diabetes. Cleve Clin J Med 84, S15-S21 (2017). J. S. Bhatti et al., Oxidative stress in the pathophysiology of type 2 diabetes and related complications: Current therapeutics strategies and future perspectives. Free Radic Biol Med 184, 114-134 (2022). K. I. Stanford, L. J. Goodyear, Exercise and type 2 diabetes: molecular mechanisms regulating glucose uptake in skeletal muscle. Adv Physiol Educ 38, 308-314 (2014). M. Flockhart et al., Excessive exercise training causes mitochondrial functional impairment and decreases glucose tolerance in healthy volunteers. Cell Metab 33, 957-970 e956 (2021). P. S. Brady, L. J. Brady, D. E. Ullrey, Selenium, vitamin E and the response to swimming stress in the rat. J Nutr 109, 1103-1109 (1979). C. J. Dillard, R. E. Litov, W. M. Savin, E. E. Dumelin, A. L. Tappel, Effects of exercise, vitamin E, and ozone on pulmonary function and lipid peroxidation. J Appl Physiol Respir Environ Exerc Physiol 45, 927-932 (1978). T. Ashton et al., Electron spin resonance spectroscopic detection of oxygen-centred radicals in human serum following exhaustive exercise. Eur J Appl Physiol Occup Physiol 77, 498-502 (1998). S. K. Powers, M. J. Jackson, Exercise-induced oxidative stress: cellular mechanisms and impact on muscle force production. Physiol Rev 88, 1243-1276 (2008). S. K. Powers, W. B. Nelson, M. B. Hudson, Exercise-induced oxidative stress in humans: cause and consequences. Free Radic Biol Med 51, 942-950 (2011). J. H. Traverse et al., Measurement of myocardial free radical production during exercise using EPR spectroscopy. Am J Physiol Heart Circ Physiol 290, H24532458 (2006). J. D. Watson, Type 2 diabetes as a redox disease. Lancet 383, 841-843 (2014). R. Rahimi, S. Nikfar, B. Larijani, M. Abdollahi, A review on the role of antioxidants in the management of diabetes and its complications. Biomed Pharmacother 59, 365-373 (2005). M. Wu et al., Liver-targeted Nano-MitoPBN normalizes glucose metabolism by improving mitochondrial redox balance. Biomaterials 222, 119457 (2019). 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. M. Wu et al., Compartmentally scavenging hepatic oxidants through AMPK/SIRT3-PGC1alpha axis improves mitochondrial biogenesis and glucose catabolism. Free Radic Biol Med 168, 117-128 (2021). J. O. Holloszy, Exercise-induced increase in muscle insulin sensitivity. J Appl Physiol (1985) 99, 338-343 (2005). J. O. Holloszy, J. Schultz, J. Kusnierkiewicz, J. M. Hagberg, A. A. Ehsani, Effects of exercise on glucose tolerance and insulin resistance. Brief review and some preliminary results. Acta Med Scand Suppl 711, 55-65 (1986). J. S. Greiwe et al., Effects of endurance exercise training on muscle glycogen accumulation in humans. J Appl Physiol (1985) 87, 222-226 (1999). B. Viollet et al., Activation of AMP-activated protein kinase in the liver: a new strategy for the management of metabolic hepatic disorders. J Physiol 574, 41-53 (2006). J. W. Zmijewski et al., Exposure to hydrogen peroxide induces oxidation and activation of AMP-activated protein kinase. J Biol Chem 285, 33154-33164 (2010). Y. Ren et al., Oxidative stress-mediated AMPK inactivation determines the high susceptibility of LKB1-mutant NSCLC cells to glucose starvation. Free Radic Biol Med 166, 128-139 (2021). S. A. Hawley et al., Use of cells expressing gamma subunit variants to identify diverse mechanisms of AMPK activation. Cell Metab 11, 554-565 (2010). K. Dong et al., Glutaredoxins concomitant with optimal ROS activate AMPK through S-glutathionylation to improve glucose metabolism in type 2 diabetes. Free Radical Biology and Medicine 101, 334-347 (2016). L. Parker, T. A. McGuckin, A. S. Leicht, Influence of exercise intensity on systemic oxidative stress and antioxidant capacity. Clin Physiol Funct Imaging 34, 377-383 (2014). F. Qin et al., Maximum oxygen consumption and quantification of exercise intensity in untrained male Wistar rats. Sci Rep 10, 11520 (2020). A. Panday, M. K. Sahoo, D. Osorio, S. Batra, NADPH oxidases: an overview from structure to innate immunity-associated pathologies. Cell Mol Immunol 12, 5-23 (2015). C. Burdon, C. Mann, T. Cindrova-Davies, A. C. Ferguson-Smith, G. J. Burton, Oxidative stress and the induction of cyclooxygenase enzymes and apoptosis in the murine placenta. Placenta 28, 724-733 (2007). M. McMahon, K. Itoh, M. Yamamoto, J. D. Hayes, Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression. J Biol Chem 278, 2159221600 (2003). M. S. Bitar, F. Al-Mulla, A defect in Nrf2 signaling constitutes a mechanism for cellular stress hypersensitivity in a genetic rat model of type 2 diabetes. Am J Physiol Endocrinol Metab 301, E1119-1129 (2011). T. Jiang et al., The protective role of Nrf2 in streptozotocin-induced diabetic nephropathy. Diabetes 59, 850-860 (2010). V. Krajka-Kuzniak, J. Paluszczak, W. Baer-Dubowska, The Nrf2-ARE signaling pathway: An update on its regulation and possible role in cancer prevention and treatment. Pharmacol Rep 69, 393-402 (2017). A. V. Budanov, J. H. Lee, M. Karin, Stressin' Sestrins take an aging fight. EMBO Mol Med 2, 388-400 (2010). M. P. Murphy et al., Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat Metab 4, 651-662 (2022). 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. H. Ahsan, 3-Nitrotyrosine: A biomarker of nitrogen free radical species modified proteins in systemic autoimmunogenic conditions. Hum Immunol 74, 1392-1399 (2013). C. M. Wong, L. Marcocci, L. Liu, Y. J. Suzuki, Cell signaling by protein carbonylation and decarbonylation. Antioxid Redox Signal 12, 393-404 (2010). O. Lopez-Acosta et al., Reactive Oxygen Species from NADPH Oxidase and Mitochondria Participate in the Proliferation of Aortic Smooth Muscle Cells from a Model of Metabolic Syndrome. Oxid Med Cell Longev 2018, 5835072 (2018). S. Heumuller et al., Apocynin is not an inhibitor of vascular NADPH oxidases but an antioxidant. Hypertension 51, 211-217 (2008). D. A. Okar et al., PFK-2/FBPase-2: maker and breaker of the essential biofactor fructose-2,6-bisphosphate. Trends Biochem Sci 26, 30-35 (2001). C. Wu, D. A. Okar, C. B. Newgard, A. J. Lange, Overexpression of 6phosphofructo-2-kinase/fructose-2, 6-bisphosphatase in mouse liver lowers blood glucose by suppressing hepatic glucose production. J Clin Invest 107, 91-98 (2001). R. A. Haeusler, K. H. Kaestner, D. Accili, FoxOs function synergistically to promote glucose production. J Biol Chem 285, 35245-35248 (2010). J. Nakae, T. Kitamura, D. L. Silver, D. Accili, The forkhead transcription factor Foxo1 (Fkhr) confers insulin sensitivity onto glucose-6-phosphatase expression. J Clin Invest 108, 1359-1367 (2001). D. B. Zorov, M. Juhaszova, S. J. Sollott, Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 94, 909-950 (2014). S. Austin, J. St-Pierre, PGC1alpha and mitochondrial metabolism--emerging concepts and relevance in ageing and neurodegenerative disorders. J Cell Sci 125, 4963-4971 (2012). T. Nie, G. J. S. Cooper, Mechanisms Underlying the Antidiabetic Activities of Polyphenolic Compounds: A Review. Front Pharmacol 12, 798329 (2021). A. Movahed et al., Efficacy and Safety of Resveratrol in Type 1 Diabetes Patients: A Two-Month Preliminary Exploratory Trial. Nutrients 12, (2020). A. F. Raimundo et al., Combined effect of interventions with pure or enriched mixtures of (poly)phenols and anti-diabetic medication in type 2 diabetes management: a meta-analysis of randomized controlled human trials. Eur J Nutr 59, 1329-1343 (2020). D. Garcia, R. J. Shaw, AMPK: Mechanisms of Cellular Energy Sensing and Restoration of Metabolic Balance. Mol Cell 66, 789-800 (2017). B. B. Zhang, G. Zhou, C. Li, AMPK: an emerging drug target for diabetes and the metabolic syndrome. Cell Metab 9, 407-416 (2009). M. Foretz, B. Guigas, B. Viollet, Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat Rev Endocrinol 15, 569-589 (2019). I. Pernicova, M. Korbonits, Metformin--mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol 10, 143-156 (2014). R. J. Shaw et al., The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 310, 1642-1646 (2005). G. Zhou et al., Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108, 1167-1174 (2001). K. Dong et al., ROS-mediated glucose metabolic reprogram induces insulin resistance in type 2 diabetes. Biochemical and biophysical research communications 476, 204-211 (2016). X. Luan et al., Exercise as a prescription for patients with various diseases. J Sport Health Sci 8, 422-441 (2019). 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. J. P. Kirwan, T. P. Solomon, D. M. Wojta, M. A. Staten, J. O. Holloszy, Effects of 7 days of exercise training on insulin sensitivity and responsiveness in type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 297, E151-156 (2009). M. A. Rogers et al., Improvement in glucose tolerance after 1 wk of exercise in patients with mild NIDDM. Diabetes Care 11, 613-618 (1988). P. A. Hansen, L. A. Nolte, M. M. Chen, J. O. Holloszy, Increased GLUT-4 translocation mediates enhanced insulin sensitivity of muscle glucose transport after exercise. J Appl Physiol (1985) 85, 1218-1222 (1998). C. E. Xirouchaki et al., Skeletal muscle NOX4 is required for adaptive responses that prevent insulin resistance. Sci Adv 7, eabl4988 (2021). K. A. Coughlan, R. J. Valentine, N. B. Ruderman, A. K. Saha, AMPK activation: a therapeutic target for type 2 diabetes? Diabetes Metab Syndr Obes 7, 241-253 (2014). J. A. Hawley, M. Hargreaves, M. J. Joyner, J. R. Zierath, Integrative biology of exercise. Cell 159, 738-749 (2014). R. C. Camacho, E. P. Donahue, F. D. James, E. D. Berglund, D. H. Wasserman, Energy state of the liver during short-term and exhaustive exercise in C57BL/6J mice. Am J Physiol Endocrinol Metab 290, E405-408 (2006). S. O. Warner, M. V. Yao, R. L. Cason, J. J. Winnick, Exercise-Induced Improvements to Whole Body Glucose Metabolism in Type 2 Diabetes: The Essential Role of the Liver. Front Endocrinol (Lausanne) 11, 567 (2020). E. C. Hinchy et al., Mitochondria-derived ROS activate AMP-activated protein kinase (AMPK) indirectly. J Biol Chem 293, 17208-17217 (2018). D. Shao et al., A redox-dependent mechanism for regulation of AMPK activation by Thioredoxin1 during energy starvation. Cell Metab 19, 232-245 (2014). M. M. Mihaylova, R. J. Shaw, The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol 13, 1016-1023 (2011). M. M. Mihaylova et al., Class IIa histone deacetylases are hormone-activated regulators of FOXO and mammalian glucose homeostasis. Cell 145, 607-621 (2011). Z. Radak et al., Exercise, oxidants, and antioxidants change the shape of the bellshaped hormesis curve. Redox Biol 12, 285-290 (2017). M. Ristow et al., Antioxidants prevent health-promoting effects of physical exercise in humans. Proc Natl Acad Sci U S A 106, 8665-8670 (2009). T. G. Bedford, C. M. Tipton, N. C. Wilson, R. A. Oppliger, C. V. Gisolfi, Maximum oxygen consumption of rats and its changes with various experimental procedures. J Appl Physiol Respir Environ Exerc Physiol 47, 1278-1283 (1979). R. E. Shepherd, P. D. Gollnick, Oxygen uptake of rats at different work intensities. Pflugers Arch 362, 219-222 (1976). 778 779 Figure legend: 780 781 Fig. 1 A model of the different mechanisms of exercise and antioxidant intervention in diabetes. 782 783 784 A graphical abstract of this study. Moderate exercise upregulated compensatory antioxidant capability and reached a high-level redox balance, whereas antioxidant intervention achieved a low-level redox balance by inhibiting oxidative stress for treating diabetes. 785 786 787 Fig. 2 Moderate exercise induced ROS production in exercise group and increased the antioxidant status. 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 A. Experimental design. T2DM rats model was fed by high-fat diet plus a low dose of STZ injection (35 mg/kg). The high-fat diet (HFD, 60% calories from fat) was started from the 1st week to the 8th week. The exercise intervention was started from 1st week to 4th week. BD. Representative protein level and quantitative analysis of NOX4 (27 kDa), COX2 (17 kDa) and Actin (45 kDa) in the rats in the Ctl, T2D and T2D + CE groups. E-G. Representative protein level and quantitative analysis of Nrf2(97 kDa), Sestrin2 (56 kDa) and Actin (45 kDa) in the rats in the Ctl, T2D and T2D + CE groups. H-K. Representative protein level and quantitative analysis of PRX1 (27 kDa), Grx1 (17 kDa), Trx1 (12 kDa) and Actin (45 kDa) in the rats in the Ctl, T2D and T2D + CE groups. The rat livers were homogenized by 1% SDS and analyzed by Western blots with the appropriate antibodies. L-M. Representative protein level and quantitative analysis of 3-NT and Actin (45 kDa) in the rat in the Ctl, T2D and T2D + CE groups. N-O. Liver protein carbonylation (O) and MDA content (P) level was detected in the rats of Ctl, T2D, T2D + CE groups. (ns: not significant; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 compared with all groups by one-way ANOVA and Tukey’s post hoc test; data are expressed as the mean ± SEM; n = 4-8 per group). 804 Figure 2_source data_01 805 806 Figure 2_source data_02 807 808 Fig. 3 Antioxidant intervention alleviates blood glucose through promoting the upregulation of reducing levels 809 810 811 812 813 814 815 816 817 818 819 820 A. Experimental design. T2DM rats model was fed by high-fat diet plus a low dose of STZ injection (35 mg/kg). The apocynin intervention was started from 1st week to 4th week. B-E. Liver protein carbonylation, MDA content (C) and TAOC (D) level were detected in the rats of Ctl, T2D and T2D + APO groups. E-H. Representative protein level and quantitative analysis of Nrf2 (97 kDa), Sestrin2 (57 kDa), Glut2 (60-70 kDa) and HSP90 (90 kDa) in the rats in the Ctl, T2D and T2D + APO groups. I. Postprandial blood glucose levels of Ctl, T2D, T2D + CE and T2D + APO groups at the end of 8th week. J. Fasting blood glucose levels of Ctl, T2D, T2D + CE and T2D + APO groups at the end of 8th week. K. Blood glucose level after oral glucose administration (0 min, 60min and 120 min) in Ctl, T2D, T2D + CE and T2D + APO groups at the end of 8th week (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 compared with all groups by one-way ANOVA and Tukey’s post hoc test; data are expressed as the mean ± SEM; n = 4-8 per group). 821 Figure 3_source data_01 822 Figure 3_source data_02 823 824 825 826 827 Fig. 4 Moderate exercise-generated ROS promotes activation of AMPK by phosphorylation and reduces blood glucose level, while excessive exercise- generated oxidative stress reduces AMPK expression and exacerbates diabetes. 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 A. Postprandial blood glucose levels of Ctl, T2D, T2D + CE, T2D + IE and T2D + EE groups at the end of 8th week. B. Blood glucose level after oral glucose administration in Ctl, T2D, T2D + CE, T2D + IE and T2D + EE groups at the end of 8th week. C-E. Representative protein level and quantitative analysis of NOX4 (27 kDa), COX2 (17 kDa) and HSP90 (90 kDa) in the rats in the Ctl, T2D, T2D + CE, T2D + IE and T2D + EE groups. F-J. Representative protein level and quantitative analysis of Ace-SOD2 (27 kDa), SOD2 (17 kDa), Grx1 (17 kDa), Trx1 (12 kDa) and Actin (45 kDa) in the rats in the Ctl, T2D, T2D + CE, T2D + IE and T2D + EE groups. K-L. Liver protein carbonylation content (k), liver MDA content (L) and AMP/ATP ratio (M) were detected in the rats of Ctl, T2D, T2D + CE, T2D + IE and T2D + EE groups. N-O. Representative protein level and quantitative analysis of P-AMPK (67 kDa), AMPK (67 kDa) and Actin (45 kDa) in the rats in the Ctl, T2D, T2D + CE, T2D + IE and T2D + EE groups. (ns: not significant; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 compared with all groups by one-way ANOVA and Tukey’s post hoc test; data are expressed as the mean ± SEM; n = 4-8 per group). 843 Figure 4_source data_01 844 Figure 4_source data_02 845 846 847 Fig. 5 Moderate exercise promoted glycolysis and mitochondrial tricarboxylic acid cycle and inhibited the gluconeogenesis in the liver of diabetic rats. 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 A-B. Representative protein level and quantitative analysis of P-PFK2 (64 kDa), PFK2 (64 kDa) and GAPDH (37 kDa) in the rats in the Ctl, T2D, T2D + CE and T2D + IE groups. C. Liver glucose level after oral glucose administration in Ctl, T2D, T2D + CE and T2D + IE groups at the end of 8th week. D. Relative concentrations of substrates for glycolysis (DHAP and Lactate) and the tricarboxylic acid cycle (citrate, succinate and malate) in the rat of Ctl, T2D, T2D + CE and T2D + IE groups. The concentration of substrates was analyzed by LC-MS/MS. E-G. Representative protein level and quantitative analysis of FoxO1 (82 kDa), GLUT2 (60-70 kDa) and Actin (45 kDa) in the rats in the Ctl, T2D, T2D + CE and T2D + IE groups. H-I. Expression of hepatic Pepck and G6C mRNA in the Ctl, T2D, T2D + CE and T2D + IE groups were evaluated by real-time PCR analysis. Values represent mean ratios of Pepck and G6pase transcripts normalized to GAPDH transcript levels. J. Schematic diagram illustrating the effect of CE and IE on glycolysis, gluconeogenesis and mitochondrial tricarboxylic acid cycle (ns: not significant; *P < 0.05, **P < 0.01, ****P < 0.0001 compared with all groups by one-way ANOVA and Tukey’s post hoc test; data are expressed as the mean ± SEM; n = 6-8 per group). 863 Figure 5_source data_01 864 Figure 5_source data_02 865 866 867 Fig. 6 Moderate exercise inhibited hepatic mitophagy, while excessive exercise exhibited opposite effect and inhibited the mitochondrial biogenesis. 868 869 870 871 872 873 874 875 A-F. Representative protein level and quantitative analysis of MFN1 (82 kDa), ATG5 (55 kDa), FIS (25 kDa), LC3A/B (14,16 kDa), PGC-1α (130 kDa) and Actin (45 kDa) in the rats in the Ctl, T2D, T2D + CE, T2D + IE and T2D + EE groups. G. TEM analysis of the ultrastructure of hepatocytes in the rats in the T2D, T2D + CE, T2D + IE and T2D + EE groups (The yellow arrows point to mitochondria). (Scale bar = 2 μm; ns: not significant; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 compared with all groups by oneway ANOVA and Tukey’s post hoc test; data are expressed as the mean ± SEM; n = 8 per group). 876 Figure 6_source data_01 877 Figure 6_source data_02 878 879 Fig. 7 Moderate ROS activates AMPK through GRX-mediated glutathionylation 880 881 882 883 884 885 886 887 888 889 890 891 892 893 A-B. Analysis of superoxide (A) and ROS (B) generation using hydroethidine (A) and H2DCFDA (B) probes in primary hepatocytes under H2O2 stress (50–200 μmol/l, 30 min). The fluorescence intensity was detected by flow cytometry. C-D. Representative protein level and quantitative analysis of GSS-adduct protein and GAPDH in primary hepatocytes under H2O2 stress (50–200 μmol/l). Hepatocytes loaded with EE-GSH-biotin were incubated with/without H2O2 for 30 min, and the amounts of GSS-protein adduct formation were determined using non-reducing SDS-PAGE and Western blot analysis with streptavidin-HRP. E-F. Representative protein level and quantitative analysis of P-AMPK (67 kDa) and AMPK (67 kDa) in primary hepatocytes under H2O2 stress (50–200 μmol/l, 30 min). G-H. AMPK cysteine gel shift immunoblot. Cysteine dependent shifts by incubation of AMPK protein with glutathione reductase and PEG-Mal. PEG2-mal labelled glutathionylation modification shifts AMPK by ~10 kDa above the native molecular weight. Representative protein level and quantitative analysis of GSS-AMPK (72 kDa), AMPK (67 kDa) and GAPDH in primary hepatocytes under H2O2 stress. 894 Figure 7_source data_01 895 Figure 7_source data_02 896 897 Fig. 8 Schematic diagram of redox balance threshold. 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 A. The increased ROS and/or decreased antioxidative capacity (AOD) cause an imbalanced redox state and declined AMPK activity in diabetic individuals. Moderate exercise promotes the activity of antioxidant enzymes by generating benign ROS to reach redox balance, and directly promotes AMPK signaling, thus reducing glucose levels in the blood and liver. Excessive exercise causes excess ROS and exceeds the redox balance threshold, inhibiting AMPK activity and expression, thus leading to exacerbation of diabetes. (AMPK and P-AMPK in grey circles indicate decrease, red circles indicate increase, blue circles indicate no significant changes). B. Dose-response curve of diabetic individuals with exercise and antioxidant intervention. The state of diabetic individuals is applicable to the description of a S-shaped curve, due to the high level of oxidative stress and decreased reduction level in diabetic individuals. With the increase of ROS, the physiological function of diabetic individuals gradually decreases. Moderate exercise shifts the S-shaped curve upward and to the right, forming a bell-shaped dose-response curve, thus reducing the sensitivity to oxidative stress in diabetic individuals and restoring redox homeostasis. However, with excessive exercise, ROS production increases beyond the threshold range of redox balance, resulting in decreased physiological function. The ROS at the peak of the bell-shaped curve for antioxidant interventions (optimal physiological activity) is lower 915 916 917 than the ROS at the peak for moderate exercise. The intervals on either side of the peak correspond to the range of redox balance thresholds, where antioxidant interventions are at low levels of redox balance and exercise is at high levels of redox balance. 918 919 Supplementary file 1a Antibody information 920 Supplementary file 1b Chow and HFD diet nutrition composition Gr aphi cabst r act Protein Carbonyl 0.25 ns 0.20 0.15 0.10 0.05 0.00 O D +C E C TL T2 T2 D D +C E 0 T2 1 D 2 2.5 2.0 1.5 1.0 0.5 Relative protein level GRX1 T2 0 J TL 1 Sestrin2 T T2 2D D +C E Actin TL T2 T2 D D +C E C 0 C 2 N MDA (mmol/l) 3-NT Level TL 3 Relative protein level G C 0.6 1 D +C E 0.9 2 T2 1.2 PRX1 GRX1 TRX1 Relative protein level D+ CE T2 D T2 Ct l COX2 D Nrf2 T2 0.8 D +C E D T2 1.0 3 T2 3 TL 1.2 Relative protein level D TL M C TL T2 T2 D D +C E C NOX4 C Actin Relative protein level Sestrin2 1.5 C TL T T2 2D D +C E F Relative protein level E T T2 2D D +C E C TL T T2 2D D +C E D+ CE T2 Relative protein level Actin 1.4 TL L CE Nrf2 Relative protein level COX2 D+ NOX4 T2 D CE D+ T2 D T2 C C Actin T2 Ct l D T2 Ct l B Relative protein level 3-NT l Ct A H I 0.0 1.6 K 1.6 5 MDA 4 3 2 1 PRX1 1.4 ns 1.2 1.0 0.8 0.6 TRX1 1.4 1.2 1.0 0.8 0.6 A Apocynin Intervention Blood glucose monitoring 4th week STZ 35mg/kg Day 0 8th week Experimental day HFD diet 0.20 6 8 D D T2 16 PO +A E D T2 T2 D +C D tl 0 0 PO Ctl T2D T2D+CE T2D+APO 10 T2 0 T2 D T2 +CE D +A PO 4 tl T2 D 10 OGTT 30 20 8 C nmol/ml 20 K T2 T2 Fasting BG 12 30 C mmol/l 40 J Blood glucose (mmol/L) Postprandial BG 0.0 D +A +A PO 0.4 0.5 D 0.6 1.0 D +A 0.8 1.5 T2 1.0 2.0 tl 1.2 T2 D T2 D 0.4 ns GLUT2 C 0.6 Relative protein level 0.8 1.4 T2 D 1.0 H Sestrin C tl Relative protein level 1.2 PO tl T2 T2 1.4 C tl HSP90 C tl C D T2 GLUT2 Relative protein level Sestrin2 G Nrf2 +A 0 D 0 PO 2 +A T2 D+ T2 D T2 l AP O F Nrf2 4 2 PO D tl C E 6 +A 0.10 4 D 0.15 IU/ml 10 nmol/l IU/ml 8 0.00 I TAOC 0.25 0.05 Ct D MDA T2 C Protein Carbonyl PO B 0 60 120 Time (minutes) 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 ns ns F 1.6 ns 1.0 0.8 0.6 I AMP/ATP P-AMPK AMPK Actin TL T T2 2D D + T2 CE D T2 +IE D +E E C T2D T2D Ctl T2D CE IE EE Ace-SOD2 SOD2 GRX1 TRX1 Actin TRX1 J 1.6 1.4 0.4 1.2 0.3 1.0 0.8 0.6 M ns T2D Ctl T2D CE IE EE EE NOX4 1.2 COX2 1.0 0.8 Actin 0.6 G Relative protein level C Protein Carbonyl content ns ns 0.2 0.1 0.0 N 8 6 T2 T2D D + T2 CE D T2 +IE D +E E COX2 TL 0 C 0 MDA (nmol/ml) 10 TL T T2 2D D + T2 CE D T2 +IE D +E E C 10 Relative protein level TL T T2 2D D + T2 CE D T2 +IE D +E E 20 IE TL T T2 2D D + T2 CE D T2 +IE D +E E GRX1 30 Ctl T2D CE Relative protein level L 40 TL T T2 2D D + T2 CE D T2 +IE D +E E 1.2 C 20 mmol/l 30 OGTT-2H C 1.4 B mmol/l E C TL T2 T2 D D + T2 CE D T2 +IE D +E E 1.8 1.6 1.4 1.2 1.0 0.8 0.6 0.4 Relative protein level T2 T2D D + T2 CE D T2 +IE D +E E TL C Bood glucose ns C T2 T2D D + T2 CE D T2 +IE D +E E TL H C Relative protein level TL T T2 2D D + T2 CE D T2 +IE D +E E C mmol/l 40 Ratio C TL T T2 2D D + T2 CE D T2 +IE D +E E Relative protein level A D NOX4 1.6 1.4 Aceyl-SOD/SOD 1.2 1.0 0.8 0.6 0.4 K MDA 6 ns 5 4 3 2 P-AMPK/Actin ns ns 4 2 0 B P-PFK2/PFK2 Liver glucose ns 1.2 1.0 0.8 0.6 OAA 1.4 J 5 Fold change 4 3 2 1 C TL T2 T2 D D +C T2 E D +I E 0 I G6PC-mRNA 2.5 Fold change 2.0 1.5 1.0 0.5 T2 T2 D D +C T2 E D +I E C TL 0.0 tl T2 D T2 C E +I D +C E 0 T2 E +I E PEPCK1-mRNA D +C D T2 H T2 D T2 C TL 0.6 1 D Actin 2 D 0.8 3 T2 GLUT2 α-KG Malate 4 T2 1.0 Glutamate Succinate GLUT2 5 1.2 FoxO1 C G FoxO1 Relative protein level Ctl T T2 2D D +C T2 E D +I E T T2 2D D +C T2 E D +I E TL C F T2D T2D CE IE Citrate 0 Relative protein level E Ace-CoA 1 TL GAPDH Lactate TL P-PFK2 DHAP 2 D +C T2 E D +I E Relative protein level 1.4 PFK2 1 1.5 2 3 1.6 C T2D T2D CE IE Ctl D C Relative content A T2 G 2 μm ns ns 1.0 0.5 0.0 T2D 2 μm E LC3 2.5 2.0 1.5 1.0 0.5 0.0 T2D+IE 2 μm TL T2 T2 D D +C T2 E D T2 +IE D +E E PGC-1α 2.0 1.5 1.0 Relative protein level MFN1 C LC3A/B T T2 2D D +C T2 E D T2 +IE D +E E B 0.5 1.6 C TL T2 T2 D D +C T2 E D T2 +IE D +E E TL Fis Relative protein level Actin C ATG5 Relative protein level MFN1 T2 D D +C T2 E D T2 +IE D +E E 2.0 T2 Fis TL D Relative protein level Ctl T2D CE IE EE C 1.5 D +C T2 E D T2 +IE D +E E D T2 C TL Relative protein level A T2D C ATG5 2.0 1.5 1.0 0.5 0.0 F PGC-1a ns 1.2 0.8 0.4 T2D+CE T2D+EE 2 μm D C Ctl H2O2 (μM) 50 100 200 GSS-protein Ctl 50 μM GSS-protein Relative protein level A 2.5 2.0 1.5 1.0 0.5 GAPDH 100 μM H2O2 (μM) E H2O2 (μM) Dihydroethidium Ctl B Relative protein level F 200 μM CTL 50 100 200 50 100 200 P-AMPK Ctl 2.5 2.0 1.5 1.0 0.5 AMPK P-AMPK/AMPK CTL 50 100 200 H2O2 (μM) G 50 μM H2O2 (μM) Ctl GSS-AMPK 100 μM AMPK 200 μM GAPDH DCF 50 100 200 Relative protein level H 2.5 GSS-AMPK 2.0 1.5 1.0 0.5 CTL 50 100 200 H2O2 Redox bal ance