AuPS Logo Contents
Previous Next PDF

Potential role of nitric oxide in contraction-stimulated glucose uptake and mitochondrial biogenesis in skeletal muscle

Glenn K. McConell and Glenn D. Wadley
Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia

Summary

1. This review will discuss the potential role of nitric oxide (NO) in the (i) regulation of skeletal muscle glucose uptake during exercise and (ii) the activation of mitochondrial biogenesis after exercise.

2. We have shown in humans that local infusion of a NO synthase (NOS) inhibitor during exercise attenuates increases in skeletal muscle glucose uptake without influencing blood flow. Recent studies from our laboratory in rodents support these human findings although rodent studies from other laboratories have yielded conflicting results.

3. There is clear evidence that NO increases mitochondrial biogenesis in non-contracting cells and also that NO influences basal skeletal muscle mitochondrial biogenesis. There have, however, been few studies examining the potential role of NO in the activation of mitochondrial biogenesis following an acute bout of exercise or in response to exercise training. Early indications are that NO is not involved in regulating the increase in mitochondrial biogenesis that occurs in response to exercise.

4. Exercise is considered the best prevention and treatment option for diabetes, but unfortunately, many people with diabetes do not or can not exercise regularly. Alternatives therapies are therefore critical to effectively manage diabetes. If skeletal muscle NO is found to play an important role in regulating glucose uptake and/or mitochondrial biogenesis, pharmaceutical agents designed to mimic these exercise effects may improve glycaemic control.

Skeletal muscle glucose uptake during exercise

Skeletal muscle accounts for over 80% of whole body insulin-stimulated glucose uptake. People with type 2 diabetes have normal levels of the Glucose Transporter GLUT-4 in their skeletal muscles but GLUT-4 translocation and glucose uptake in response to insulin is reduced.1 Importantly, however, skeletal muscle GLUT-4 translocation2 and glucose uptake during exercise is normal in people with diabetes.3 Indeed, blood glucose levels can decrease to normal levels during 45 min of intense exercise in people with type 2 diabetes.4

The signalling pathway(s) associated with contraction-stimulated glucose uptake are not fully understood, but are known to differ from insulin pathways. For example wortmannin, a phosphatidylinositol 3-kinase (PI3 kinase) inhibitor, blocks insulin-stimulated glucose uptake in skeletal muscle but it has no effect on contraction-stimulated glucose uptake. Possible candidates regulating glucose uptake during exercise include calcium-calmodulin dependent protein kinase II (CaMKII), protein kinase C, AMP-activated protein kinase (AMPK), reactive oxygen species and NO5-7 (See Figure 1). It is likely that more than one regulator is involved in the control of skeletal muscle glucose uptake during exercise, and that some redundancy exists. The first half of this review will focus on the potential role of NO/NO synthase (NOS) in contraction-stimulated glucose uptake in skeletal muscle.

Figure 1

Figure 1. The potential regulation and regulating pathways of skeletal muscle glucose uptake during contraction/exercise with a focus on nitric oxide. Superoxide (O2·), Nitric oxide (NO·), neuronal isoform of NO synthase (nNOS), calcium-calmodulin dependent protein kinase II (CaMKII), Protein kinase C (PKC), hydrogen peroxide (H2O2), AMP-activated protein kinase (AMPK), protein kinase G (PKG), cyclic guanosine monophosphate (cGMP).

Does NO/NOS regulate contraction-stimulated glucose uptake?

The production of NO is catalysed by NOS enzymes which convert L-arginine to L-citrulline and NO. NOS activity increases in skeletal muscle during in situ contractions8 and during in vivo exercise in rats9 (Figure 1). The resulting increase in NO release from contracting muscle10 and contracting cells11 can be prevented by NOS inhibition.8-10 NO binds to the haem group of soluble guanylate cyclase (sGC), producing the second messenger cyclic guanosine monophospate (cGMP) (Figure 1). cGMP increases during in vitro muscle contractions in mice and this increase in cGMP is prevented by NOS inhibition.12

In skeletal muscle, neuronal (n) NOS is the primary source of NO production during contraction.11-13 Indeed, skeletal muscle cGMP concentration increases during contraction in endothelial (e) NOS knock-out mice but not nNOS knock-out mice.12 There is very little expression of inducible (i) NOS in normal skeletal muscle.14,15 We found that infusion of the NOS inhibitor NG-monomethyl-L-arginine (L-NMMA) in to the femoral artery during cycling exercise substantially attenuated the increase in leg glucose uptake in healthy young individuals6 and especially in people with type 2 diabetes.3 Importantly, local infusion of the NOS inhibitor had no effect on leg blood flow, arterial blood pressure, or insulin and glucose concentrations during exercise.3,6 These studies suggest that NO is required for normal increases in glucose uptake during exercise in humans. We also found that infusion of the NOS precursor L-arginine (0.5 g/min) during prolonged exercise in healthy young men significantly increased glucose disposal during exercise.16 It has been shown that L-arginine increases basal NO production by rat skeletal muscle in vitro10 so we assume that the L-arginine infusion increased skeletal muscle NO production during exercise.

It should be noted that in our human studies we did not examine whether exercise increased NOS activity and whether this was prevented by NOS inhibition or that NOS activity was increased by L-arginine infusion during exercise. Future studies are warranted to address this cognitive gap. We have followed up these human studies with rodent experiments which allow a more thorough examination of mechanisms. In recent preliminary findings we have indications that nNOS knock-out mice display essentially no increase in glucose uptake during in vitro contractions (Linden et al. unpublished observations) [K.C. Linden,* J.G. Ryall, C. van der Poel, J.D. Schertzer, G.S. Lynch & G.K. McConell.* 2008. *Exercise Physiology and Metabolism Laboratory, and Basic and Clinical Myology Laboratory, Department of Physiology, University of Melbourne, Victoria 3010, Australia.] and that NOS inhibition (by L-NMMA) attenuates skeletal muscle glucose uptake during in vitro contractions in C57Bl6 mice (Merry et al. unpublished observations). [T. Merry, G. Lynch, & G. McConell. 2008. Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia.] Since there is no blood flow during in vitro muscle contractions, these results confirm that the effects of a reduction/prevention of NO production (NOS inhibition or nNOS knock-out mice) on glucose uptake are independent of the potential effects of NO on blood flow. Indeed, we also found that NOS inhibition, which prevented the contraction-induced increase in skeletal muscle NOS activity, attenuated increases in glucose uptake during in situ contractions in rats without affecting muscle capillary blood flow, suggesting that NOS inhibition was affecting the skeletal muscle fibres per se.8 Taken together with our human findings, these studies suggest that NO is essential for normal increases in skeletal muscle glucose uptake during exercise.

It is worth noting that rat studies from other laboratories have yielded conflicting results (e.g. Higaki et al., 2001,17 Roberts et al., 199718). For example, two studies found NOS inhibition prevented increases in contraction-stimulated glucose uptake5,18 but two other studies reported no effect of NOS inhibition on contraction-stimulated glucose uptake.17,19 In general, the rodent studies in this area of inquiry have been difficult to interpret, since in these studies, unlike in our human studies and our recent rodent studies where glucose uptake is measured during exercise, glucose transport/uptake measurements were usually made 20 or more minutes after contractions or treadmill exercise. The potential reasons for these contradictory findings in the studies using rodents have been discussed elsewhere.20

Downstream signalling of NOS/NO in terms of glucose uptake

It is important to examine the potential mechanisms of how NO increases glucose uptake during contractions. To date, the only studies examining phenomena downstream of NOS in relation to glucose uptake were conducted in non-contracting muscles.21,22 Clearly, one can not assume that the regulation of muscle glucose uptake at rest is similar to the regulation during exercise.

In non-contracting isolated skeletal muscles, NO appears to exert its effects by modulating cGMP levels.21,22 Increases in NO stimulate soluble guanylate cyclase, producing cGMP (Figure 1). In isolated skeletal muscles from mice and frogs, contraction increases muscle cGMP levels and the cGMP analogue 8-bromo-cGMP increases glucose uptake in isolated rat muscles.21,22 In addition, NO donors such as sodium nitroprusside (SNP) raise skeletal muscle cGMP content17,21,22 and increase glucose uptake, presumably as a result of the activation of sGC.21,22 Indeed, addition of LY-83583, an inhibitor of sGC, decreases skeletal muscle cGMP content and reduces basal 2-deoxyglucose transport in isolated rat muscle to a similar extent as with L-NMMA.22 LY-83583 also completely abolishes SNP augmented glucose transport.22 However, LY-83583, in addition to inhibiting sGC, also inhibits nNOS.23 It is therefore important that the more specific sGC inhibitor 1H-[1,2,4]Oxadiazole[4,3-a]quinoxalin-1-one (ODQ) be used24 and also that contraction studies be conducted. It is also important that studies examine whether cGMP-dependent protein kinase (PKG) activity is increased with contraction/exercise (Figure 1).

It has become clear that in many cell types NO also signals through cGMP-independent mechanisms, especially S-nitrosylation,25,26 the covalent addition of NO to a cysteine thiol/sulfhydryl group, resulting in S-nitrosothiols (Figure 1). S-nitrosylation is “increasingly becoming recognized as a ubiquitous regulatory reaction comparable to phosphorylation”.26 Indeed, it has been demonstrated in vascular smooth muscle cells, human endothelial cells, intact hearts and in skeletal muscle that S-nitrosylation is involved in cGMP-independent signalling effects of NO27-30 and that proteins associated with glucose transport regulation (e.g. Akt/PKB) are susceptible to S-nitrosylation in skeletal muscle.30 It is important that studies examine whether NO increases glucose uptake during contraction/exercise, at least in part, via increases in S-nitrosylation.

Finally, NO may exert its effects during exercise in part via tyrosine nitration by peroxynitrite.31 Peroxynitrite is formed by a non-enzymatic reaction between NO and the reactive oxygen species (ROS), superoxide (Figure 1). It has been reported that during contraction of myocytes there are increases in both superoxide and NO, and therefore peroxynitrite11 (Figure 1). Note that this study11 involved contraction in healthy muscle cells so is indicative of physiological signalling and not a pathological disease process, as may be the case with peroxynitrite. At physiological levels, peroxynitrite upregulates an array of signalling cascades, through the inhibition of phosphatases and direct activation of several different protein kinases (e.g. AMPK and MAP kinases) involved in regulating skeletal muscle glucose uptake.25,32

AMPK does not increase glucose uptake via activation of nNOS

5-aminoimidazole-4-carboxyamide-ribonucleoside (AICAR), a cell-permeable activator of AMPK, increases rat skeletal muscle glucose uptake, in vitro and in vivo. Like exercise, AICAR increases the translocation of GLUT-4 to the sarcolemma and appears to act via an insulin-independent mechanism. AMPK phosphorylates and activates eNOS33 and in the heart this plays a role in glucose uptake.34 A study in rats found that AICAR increased NOS activity and glucose uptake in muscle fibre bundles, and that these effects could be prevented by NOS inhibition. However, we have found that NOS inhibition has no effect on AICAR-activated glucose transport in isolated rat skeletal muscle.35 In addition, we found that although AMPK phosphorylates nNOS during contraction in situ in rats,8 and during exercise in human skeletal muscle,36 in vitro phosphorylation of recombinant nNOS by purified AMPK has little effect on nNOS activity (Lee-Young et al. unpublished observations). [R.S. Lee-Young,* Z.-P. Chen, B.J. Michell, G.D. Wadley,* B.E. Kemp & G.K. McConell.* 2008. *Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia, St. Vincent's Institute, Fitzroy, Victoria 3065, Australia CSIRO Molecular & Health Technologies, Parkville, Victoria 3052, Australia.] It is unlikely, therefore, that AMPK exerts effects on glucose uptake via phosphorylation of nNOS, which, as mentioned earlier, is the main isoform in muscle. Indeed, preliminary evidence indicates that AICAR increases glucose uptake normally in skeletal muscle of nNOS knock-out mice in vitro (Linden et al. unpublished observations). [op cit.]

Mitochondria, exercise and diabetes

The skeletal muscle of people with type 2 diabetes have reduced mitochondrial function, largely due to reduced mitochondrial volume.37 This reduced mitochondrial volume/function and elevated intramuscular lipids are associated with insulin resistance in skeletal muscle.38-41 However, it is still unclear if reduced mitochondrial volume/function is a cause or a consequence of insulin resistance.42,43 Some studies,39,41,44 but not all,40 have reported reduced mitochondrial synthesis (biogenesis) in insulin resistant skeletal muscle.

Endurance exercise training increases skeletal muscle mitochondrial volume45,46 and mitochondrial biogenesis.45,47-49 It is possible that increases in skeletal muscle mitochondrial biogenesis contribute to the increase in skeletal muscle insulin sensitivity observed after exercise training.50 Although this has not been extensively examined, evidence suggests that people with type 2 diabetes, who have reduced skeletal muscle mitochondrial biogenesis, respond to exercise training with a normal increase in mitochondrial enzymes and insulin sensitivity.50

Many research groups are attempting to determine how exercise increases mitochondrial biogenesis and it appears that the same signals that may be involved in regulating glucose uptake during exercise (e.g. AMPK, CaMK and NO) may also be activating gene expression of key markers of mitochondrial biogenesis such as peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1α (PCG-1α), nuclear respiratory factors 1 and 2 (NRF-1, NRF-2) and mitochondrial transcription factor A (mtTFA) after exercise.51,52 The next section will discuss the potential role of NO in basal and contraction-stimulated mitochondrial biogenesis.

Does NO/NOS play a role in mitochondrial biogenesis after exercise?

It has been shown that NO donors and analogues of the downstream messenger of NO, cGMP, increase mitochondrial biogenesis in muscle cells.53 Although nNOS is the major NOS isoform in skeletal muscle, both eNOS and nNOS isoforms are expressed within skeletal muscle fibers, with eNOS more abundant in oxidative, and nNOS more abundant in glycolytic skeletal muscles in rodents.12,54-56 NO production, total NOS activity and cGMP, increase during contraction in rodent skeletal muscle9,10,12 and may therefore be involved in the upregulation of mitochondrial biogenesis after exercise.

We found that ingestion of the non-specific NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME) for 2 days reduced basal skeletal muscle mitochondrial biogenesis but had no effect on the increase in mitochondrial biogenesis after an acute bout of exercise.57 Similarly, the increase in mitochondrial biogenesis markers in response to acute exercise and exercise training is intact in nNOS knock-out and eNOS knock-out mice.58 Therefore, these data suggest there is little involvement of NOS in the activation of mitochondrial biogenesis following exercise. Interestingly, however, rather than basal mitochondrial biogenesis being lower in nNOS knock-out mice muscle it was actually higher,58 a finding consistent with a previous report of elevated citrate synthase activity in the skeletal muscles of nNOS knock-out mice.59 The molecular mechanism(s) for elevated basal mitochondrial biogenesis in the muscle of nNOS knock-out mice is unclear, although it is independent of two key signalling proteins for mitochondrial biogenesis; phosphorylation of p38 MAPK and AMPK,58 which were normal in basal nNOS knock-out mouse skeletal muscle.

Some,53,60 but not all studies,58 have also reported reduced basal mitochondrial biogenesis in the skeletal muscles of eNOS knock-out mice. However, methodological differences, including eNOS knock-out mice being generated from different backgrounds and from differently generated eNOS gene deletions61,62 make comparisons between studies difficult. Further investigations are needed to elucidate a role for eNOS in basal mitochondrial biogenesis in skeletal muscle.

Although more research is required, these studies suggest that NO plays a role in basal mitochondrial biogenesis but not in the increase in mitochondrial biogenesis in response to exercise. Further investigations are required to determine the exact roles of the eNOS and nNOS isoforms in basal mitochondrial biogenesis.

Conclusions

There is good, albeit not fully consistent, evidence that NO is essential for normal increases in glucose uptake during contraction/exercise. Further studies are now required to determine the factors downstream of NO/nNOS that regulate contraction-stimulated glucose uptake. In contrast, although further studies are required it appears that NO/NOS are not important in the activation of skeletal muscle mitochondrial biogenesis following exercise although NO/NOS may play a role in basal skeletal muscle mitochondrial biogenesis.

Acknowledgements

We would like to acknowledge our collaborators on the studies mentioned especially Dr Scott Bradley, Associate Professor Bronwyn Kingwell, Associate Professor Stephen Rattigan, Troy Merry and Kelly Linden. We would also like to express our appreciation to the participants involved in our research and the funding from: the National Health and Medical Research Council (NHMRC) of Australia and Diabetes Australia.

References

1.  Zierath JR, He L, Guma A, Odegoard Wahlstrom E, Klip A, Wallberg-Henriksson H. Insulin action on glucose transport and plasma membrane GLUT4 content in skeletal muscle from patients with NIDDM. Diabetologia 1996; 39:1180-9.

2.  Kennedy JW, Hirshman MF, Gervino EV, et al. Acute exercise induces GLUT4 translocation in skeletal muscle of normal human subjects and subjects with type 2 diabetes. Diabetes 1999; 48: 1192-7.

3.  Kingwell B, Formosa M, Muhlmann M, Bradley S, McConell G. Nitric oxide synthase inhibition reduces glucose uptake during exercise in individuals with type 2 diabetes more than in control subjects. Diabetes 2002; 51: 2572-80.

4.  Musi N, Hayashi T, Fujii N, Hirshman MF, Witters LA, Goodyear LJ. AMP-activated protein kinase activity and glucose uptake in rat skeletal muscle. Am. J. Physiol. 2001; 280: E677-84.

5.  Balon TW, Nadler JL. Evidence that nitric oxide increases glucose transport in skeletal muscle. J. Appl. Physiol. 1997; 82: 359-63.

6.  Bradley SJ, Kingwell BA, McConell GK. Nitric oxide synthase inhibition reduces leg glucose uptake but not blood flow during dynamic exercise in humans Diabetes 1999; 48:1815-21.

7.  Richter EA, Nielsen JN, Jorgensen SB, Frosig C, Birk JB, Wojtaszewski JF. Exercise signalling to glucose transport in skeletal muscle. Proc. Nutr. Soc. 2004; 63: 211-6.

8.  Ross RM, Wadley GD, Clark MG, Rattigan S, McConell GK. Local NOS inhibition reduces skeletal muscle glucose uptake but not capillary blood flow during in situ muscle contraction in rats. Diabetes 2007; 56: 2885-92.

9.  Roberts CK, Barnard RJ, Jasman A, Balon TW. Acute exercise increases nitric oxide synthase activity in skeletal muscle. Am. J. Physiol. 1999; 277: E390-4.

10.  Balon TW, Nadler JL. Nitric oxide release is present from incubated skeletal muscle preparations. J. Appl. Physiol. 1994; 77: 2519-21.

11.  Pattwell DM, McArdle A, Morgan JE, Patridge TA, Jackson MJ. Release of reactive oxygen and nitrogen species from contracting skeletal muscle cells. Free Radical Bio. & Med. 2004; 37: 1064-72.

12.  Lau KS, Grange RW, Isotani E, et al. nNOS and eNOS modulate cGMP formation and vascular response in contracting fast-twitch skeletal muscle. Physiol. Genomics 2000; 2: 21-7.

13.  Hirschfield W, Moody MR, O'Brien WE, Gregg AR, Bryan RM, Jr., Reid MB. Nitric oxide release and contractile properties of skeletal muscles from mice deficient in type III NOS. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000; 278: R95-100.

14.  McConell GK, Bradley SJ, Stephens TJ, Canny BJ, Kingwell BA, Lee-Young RS. Skeletal muscle nNOSμ protein content is increased by exercise training in humans. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007; 293: R821-8.

15.  Rudnick J, Puttmann B, Tesch PA, et al. Differential expression of nitric oxide synthases (NOS 1-3) in human skeletal muscle following exercise countermeasure during 12 weeks of bed rest. Faseb J. 2004; 18: 1228-30.

16.  McConell GK, Huynh NN, Lee-Young RS, Canny BJ, Wadley GD. L-Arginine infusion increases glucose clearance during prolonged exercise in humans. Am. J. Physiol. 2006; 290: E60-6.

17.  Higaki Y, Hirshman MF, Fujii N, Goodyear LJ. Nitric oxide increases glucose uptake through a mechanism that is distinct from the insulin and contraction pathways in rat skeletal muscle. Diabetes 2001; 50: 241-7.

18.  Roberts CK, Barnard RJ, Scheck SH, Balon TW. Exercise-stimulated glucose transport in skeletal muscle is nitric oxide dependent. Am. J. Physiol. 1997; 273: E220-5.

19.  Etgen GJ, Fryburg DA, Gibbs EM. Nitric oxide stimulates skeletal muscle glucose transport through a calcium/contraction- and phosphatidylinositol-3-kinase-independent pathway. Diabetes 1997; 46: 1915-9.

20.  McConell GK, Kingwell BA. Does nitric oxide regulate skeletal muscle glucose uptake during exercise? Exer. & Sport Sci. Rev. 2006; 34: 36-41.

21.  Young ME, Leighton B. Fuel oxidation in skeletal muscle is increased by nitric oxide/cGMP – evidence for involvement of cGMP-dependent protein kinase. FEBS Lett 1998; 424:79-83.

22.  Young ME, Radda GK, Leighton B. Nitric oxide stimulates glucose transport and metabolism in rat skeletal muscle in vitro. Biochem. J. 1997; 322: 223-8.

23.  Luo D, Das S, Vincent SR. Effects of methylene blue and LY83583 on neuronal nitric oxide synthase and NADPH-diaphorase. Euro. J. Pharm. 1995; 290: 247-51.

24.  Garthwaite J, Southam E, Boulton CL, Nielsen EB, Schmidt K, Mayer B. Potent and selective inhibition of nitric oxide-sensitive guanylyl cyclase by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one. Mol. Pharmaco. 1995; 48: 184-8.

25.  Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007; 87: 315-424.

26.  Broillet MC. S-nitrosylation of proteins. Cell Mol. Life Sci. 1999; 55:1036-42.

27.  Paolocci N, Ekelund UE, Isoda T, et al. cGMP-independent inotropic effects of nitric oxide and peroxynitrite donors: potential role for nitrosylation. Am. J. Physiol. Heart Circ. Physiol. 2000; 279: H1982-8.

28.  White TA, Walseth TF, Kannan MS. Nitric oxide inhibits ADP-ribosyl cyclase through a cGMP-independent pathway in airway smooth muscle. Am. J. Physiol. Lung Cell Mol. Physiol. 2002; 283: L1065-71.

29.  Selemidis S, Dusting GJ, Peshavariya H, Kemp-Harper BK, Drummond GR. Nitric oxide suppresses NADPH oxidase-dependent superoxide production by S-nitrosylation in human endothelial cells. Cardio. Res. 2007; 75: 349-58.

30.  Yasukawa T, Tokunaga E, Ota H, Sugita H, Martyn JA, Kaneki M. S-nitrosylation-dependent inactivation of Akt/protein kinase B in insulin resistance. J. Biol. Chem. 2005; 280: 7511-8.

31.  Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS. Protein S-nitrosylation: purview and parameters. Nat. Rev. 2005; 6: 150-66.

32.  Zou MH, Hou XY, Shi CM, Nagata D, Walsh K, Cohen RA. Modulation by peroxynitrite of Akt- and AMP-activated kinase-dependent Ser1179 phosphorylation of endothelial nitric oxide synthase. J. Biol. Chem. 2002; 277: 32552-7.

33.  Chen ZP, Mitchelhill KI, Michell BJ, et al. AMP-activated protein kinase phosphorylation of endothelial NO synthase. FEBS Lett. 1999; 443: 285-9.

34.  Li J, Hu X, Selvakumar P, et al. Role of the nitric oxide pathway in AMPK-mediated glucose uptake and GLUT4 translocation in heart muscle. Am. J. of Physiol. 2004; 287: E834-41.

35.  Stephens TJ, Canny BJ, Snow RJ, McConell GK. 5′-aminoimidazole-4-carboxyamide-ribonucleoside-activated glucose transport is not prevented by nitric oxide synthase inhibition in rat isolated skeletal muscle. Clin. Exp. Pharmacol. Physiol. 2004; 31: 419-23.

36.  Chen ZP, McConell GK, Michell BJ, Snow RJ, Canny BJ, Kemp BE. AMPK signaling in contracting human skeletal muscle: acetyl-CoA carboxylase and NO synthase phosphorylation. Am. J. Physiol. 2000; 279: E1202-6.

37.  Boushel R, Gnaiger E, Schjerling P, Skovbro M, Kraunsoe R, Dela F. Patients with type 2 diabetes have normal mitochondrial function in skeletal muscle. Diabetologia 2007; 50: 790-6.

38.  Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 2002; 51: 2944-50.

39.  Mootha VK, Lindgren CM, Eriksson KF, et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 2003; 34: 267-73.

40.  Morino K, Petersen KF, Dufour S, et al. Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J. Clin. Invest. 2005; 115: 3587-93.

41.  Patti ME, Butte AJ, Crunkhorn S, et al. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: Potential role of PGC1 and NRF1. Poc. Natl. Acad. Sci. USA 2003; 100: 8466-71.

42.  Asmann YW, Stump CS, Short KR, et al. Skeletal muscle mitochondrial functions, mitochondrial DNA copy numbers, and gene transcript profiles in type 2 diabetic and nondiabetic subjects at equal levels of low or high insulin and euglycemia. Diabetes 2006; 55: 3309-19.

43.  Wredenberg A, Freyer C, Sandstrom ME, et al. Respiratory chain dysfunction in skeletal muscle does not cause insulin resistance. Biochem. Biophy. Res. Com. 2006; 350: 202-7.

44.  Koves TR, Li P, An J, et al. Peroxisome proliferator-activated receptor-γ co-activator 1α-mediated metabolic remodeling of skeletal myocytes mimics exercise training and reverses lipid-induced mitochondrial inefficiency. J. Biol. Chem. 2005; 280: 33588-98.

45.  Holloszy JO. Biochemical adaptations in muscle. Effects of exercise on mitochondrial oxygen uptake and respiratory enzyme activity in skeletal muscle. J. Biol. Chem. 1967; 242: 2278-82.

46.  Hoppeler H, Howald H, Conley K, et al. Endurance training in humans: aerobic capacity and structure of skeletal muscle. J. Appl. Physiol. 1985; 59: 320-7.

47.  Baar K, Wende AR, Jones TE, et al. Adaptations of skeletal muscle to exercise: rapid increase in the transcriptional co-activator PGC-1. FASEB J. 2002; 16: 1879-86.

48.  Murakami T, Shimomura Y, Yoshimura A, Sokabe M, Fujitsuka N. Induction of nuclear respiratory factor-1 expression by an acute bout of exercise in rat muscle. Biochim. Biophys. Acta. 1998; 1381: 113-22.

49.  Pilegaard H, Saltin B, Neufer PD. Exercise induces transient transcriptional activation of the PGC-1α gene in human skeletal muscle. J. Physiol. 2003; 546: 851-8.

50.  Bruce CR, Kriketos AD, Cooney GJ, Hawley JA. Disassociation of muscle triglyceride content and insulin sensitivity after exercise training in patients with Type 2 diabetes. Diabetologia 2004; 47: 23-30.

51.  Wright DC, Geiger PC, Han DH, Jones TE, Holloszy JO. Calcium induces increases in peroxisome proliferator-activated receptor γ coactivator-1α and mitochondrial biogenesis by a pathway leading to p38 mitogen activated protein kinase activation. J. Biol. Chem. 2007; 282: 18793-9.

52.  Terada S, Tabata I. Effects of acute bouts of running and swimming exercise on PGC-1α protein expression in rat epitrochlearis and soleus muscle. Am. J. Physiol. 2004; 286: E208-16.

53.  Nisoli E, Falcone S, Tonello C, et al. Mitochondrial biogenesis by NO yields functionally active mitochondria in mammals. Proc. Natl. Acad. Sci. USA 2004; 101: 16507-12.

54.  Hussain SN, El-Dwairi Q, Abdul-Hussain MN, Sakkal D. Expression of nitric oxide synthase isoforms in normal ventilatory and limb muscles. J. Appl. Physiol. 1997; 83: 348-53.

55.  Kobzik L, Stringer B, Balligand JL, Reid MB, Stamler JS. Endothelial type nitric oxide synthase in skeletal muscle fibers: mitochondrial relationships. Biochem. Biophy. Res. Com. 1995; 211: 375-81.

56.  Kapur S, Bedard S, Marcotte B, Cote CH, Marette A. Expression of nitric oxide synthase in skeletal muscle: a novel role for nitric oxide as a modulator of insulin action. Diabetes 1997; 46: 1691-700.

57.  Wadley GD, McConell GK. Effect of nitric oxide synthase inhibition on mitochondrial biogenesis in rat skeletal muscle. J. Appl. Physiol. 2007; 102: 314-20.

58.  Wadley GD, Choate J, McConell GK. NOS isoform-specific regulation of basal but not exercise-induced mitochondrial biogenesis in mouse skeletal muscle. J. Physiol. 2007; 585: 253-62.

59.  Schild L, Jaroscakova I, Lendeckel U, Wolf G, Keilhoff G. Neuronal nitric oxide synthase controls enzyme activity pattern of mitochondria and lipid metabolism. Faseb J. 2006; 20: 145-7.

60.  Le Gouill E, Jimenez M, Binnert C, et al. Endothelial nitric oxide synthase (eNOS) knockout mice have defective mitochondrial β-oxidation. Diabetes 2007; 56: 2690-6.

61.  Huang PL, Huang Z, Mashimo H, et al. Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 1995; 377: 239-42.

62.  Shesely EG, Maeda N, Kim HS, et al. Elevated blood pressures in mice lacking endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 1996; 93: 13176-81.


Received 31 March 2008, in revised form 24 May 2008. Accepted 28 May 2008.
© G.K. McConell 2008.