Você está na página 1de 7

Insulin resistance and the metabolism of branched-chain amino

acids
Jingyi Lu
1
, Guoxiang Xie
2,3
, Weiping Jia
1
, Wei Jia ()
2,3,a
1
Shanghai Diabetes Institute; Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Afliated Sixth Peoples
Hospital; Shanghai Key Laboratory of Diabetes Mellitus; Shanghai Clinical Center for Diabetes, Shanghai 200233, China;
2
Center for
Translational Medicine, Shanghai Jiao Tong University Afliated Sixth Peoples Hospital, Shanghai 200233, China;
3
Center for
Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC
28081, USA
Higher Education Press and Springer-Verlag Berlin Heidelberg 2013
Abstract Insulin resistance (IR) is a key pathological feature of metabolic syndrome and subsequently causes
serious health problems with an increased risk of several common metabolic disorders. IR related metabolic
disturbance is not restricted to carbohydrates but impacts global metabolic network. Branched-chain amino acids
(BCAAs), namely valine, leucine and isoleucine, are among the nine essential amino acids, accounting for 35% of
the essential amino acids in muscle proteins and 40% of the preformed amino acids required by mammals. The
BCAAs are particularly responsive to the inhibitory insulin action on amino acid release by skeletal muscle and
their metabolism is profoundly altered in insulin resistant conditions and/or insulin deciency. Although increased
circulating BCAA concentration in insulin resistant conditions has been noted for many years and BCAAs have
been reported to be involved in the regulation of glucose homeostasis and body weight, it is only recently that
BCAAs are found to be closely associated with IR. This review will focus on the recent ndings on BCAAs from
both epidemic and mechanistic studies.
Keywords branched-chain amino acids; leucine; isoleucine; valine; insulin resistance
Introduction
Insulin resistance (IR) is a physiological condition in which
target organs including skeletal muscle, adipose tissue and
liver fail to respond to the action of insulin effectively. IR
causes incomplete suppression of hepatic glucose output and
impaired insulin-mediated glucose uptake and utilization in
skeletal muscle and adipose tissue, leading to increased
insulin requirements. IR and abnormal insulin secretion are
two key disorders for the development of type 2 diabetes
(T2DM) [1]. In addition, IR is closely associated with
metabolic syndrome [2], which includes central obesity,
hyperglycemia, hypertension and dyslipidemia, common risk
factors for cardiovascular disease. Obesity is a well-
established risk factor for the development of IR [3]. It is
estimated that over 200 million men and nearly 300 million
women were obese worldwide in 2008 [4]. Obesity-
associated resistance to the peripheral action of insulin is a
major underlying mechanism causing the metabolic syn-
drome and ultimately T2DM. IR related metabolic distur-
bance is widespread, involving not only carbohydrate and fat
metabolism, but also protein metabolism [5]. Currently, the
mechanisms by which increased fat accumulation leads to IR
and metabolic syndrome are not completely understood.
Branched-chain amino acids (BCAAs) include leucine,
isoleucine, and valine, which are three of the nine essential
amino acids and are relatively abundant in the food supply,
accounting for 15%25% of the total protein intake [6]. In
addition to the effects on protein synthesis and degradation,
BCAAs were shown to have the capability to improve
skeletal muscle glucose uptake and whole body glucose
oxidation [79]. Cota et al. [10] demonstrated that central
administration of leucine decreases food intake and body
weight through the activation of mammalian target of
rapamycin (mTOR) in hypothalamus. Accordingly, it has
been proposed that BCAAs may be responsible for some of
the benecial effects of high-protein diets on body weight and
adiposity [6,11,12]. In fact, the elevation of blood levels of
REVIEW
Received October 9, 2012; accepted January 9, 2013
Correspondence: w_jia@uncg.edu
Front. Med. 2013, 7(1): 5359
DOI 10.1007/s11684-013-0255-5
amino acids, particularly BCAAs, in insulin resistant states of
obesity [1316] has long been noted for over 40 years.
However, BCAAs has never been recognized as a major
player in the modulation of insulin action before the study
conducted by Newgard et al. [17], in which a BCAA-related
metabolic signature showed strongest association with IR.
Thereafter, a rapidly growing number of studies linking
BCAAs with IR emerged. In this review, we will summarize
recent studies about the epidemic associations of BCAAs
with IR, possible mechanisms for their elevations in insulin
resistant states and the role of BCAAs in insulin signaling.
BCAA metabolism
The rst two steps of the pathway in BCAA catabolism by
branched-chain aminotransferase (BCAT) and the branched-
chain -keto acid dehydrogenase (BCKDH) complex are
common to the three BCAAs [1821]. BCAT isozymes
(including mitochondrial BCAT and cytosolic BCAT)
catalyze the transamination of BCAAs, which is the
reversible transfer of the -amino group to -ketoglutarate.
The BCKDH complex, which contains multiple copies of
three enzymes, a branched-chain -keto acid decarboxylase
(E1), a dihydrolipoyl transacylase (E2), and a dihydrolipoyl
dehydrogenase (E3), catalyzes the oxidative decarboxylation
of the branched-chain -keto acid products of the transamina-
tion reaction. Activity of the BCKDH complex is regulated by
phosphorylation/dephosphorylation of the E1 subunit [22].
The BCKDH kinase is responsible for inactivation of the
complex by phosphorylation of the E1 subunit of the
complex [23], and the BCKDH phosphatase is responsible for
activation of the complex by dephosphorylation of E1 [24].
In summary, the catabolism of BCAAs, valine, leucine and
isoleucine, produces three -keto acids which are further
oxidized by a common branched-chain -keto acid dehy-
drogenase, yielding three CoA derivatives. Subsequently, the
metabolic pathway diverges, producing intermediates to be
used in TCA cycle.
Linkage between BCAAs and IR
In the pioneering study conducted by Newgard et al. [17],
application of metabolomics technologies has revealed that
BCAAs and related metabolites are more strongly associated
with IR than many common lipid species. Principle
component analysis (PCA) showed that the component
comprising BCAAs, aromatic amino acids and BCAA by-
products was most strongly associated with obesity. Impor-
tantly, this component was positively related to homeostasis
model assessment-insulin resistance (HOMA-IR) index (r =
0.58, P < 0.001), an indicator of IR. Furthermore, the
authors found that feeding Wistar rats with a high-fat (HF)
diet with BCAA (HF/BCAA) reduced food intake and body
weight, but caused the animals to be equally insulin resistant
compared to heavier rats fed on a non-supplemented HF diet,
conrming the contribution of BCAAs to the development of
IR. Their ndings were further validated in a cross-sectional
study [25], in which plasma large neutral amino acids
including valine and leucine/isoleucine accounted for most of
the variability of IR in 73 overweight/obese individuals
without diabetes. Next, the association between BCAAs and
IR in normal weight subjects was also found. In a study aimed
to investigate the correlates of insulin sensitivity in two
groups of healthy individuals, multivariate analysis of the 191
metabolites measured by mass spectrometry revealed that
reduction in leucine/isoleucine along with glycerol during
oral glucose tolerance test represents the strongest predictor
of insulin sensitivity and the decrease of plasma BCAA levels
is blunted in insulin-resistant subjects during the oral glucose
tolerance test [26]. In 263 non-obese Asian-Indian and
Chinese men, Tai et al. [27] found that fatty acids and
inammatory cytokines, which are commonly regarded as IR
risk factors, could not discriminate between insulin sensitive
and insulin resistant subjects, whereas IR was signicantly
associated with the increased levels of alanine, proline,
valine, leucine/isoleucine, phenylalanine, tyrosine, glutamate/
glutamine and ornithine.
In addition to cross-sectional observations, prospective
studies demonstrated that the alterations of BCAAs may
presage the onset of overt diabetes. A study in the
Framingham cohort conducted by Wang et al. [28] reported
that baseline BCAAs and aromatic amino acids were
signicantly associated with future diabetes, and a combina-
tion of three amino acids (isoleucine, phenylalanine and
tyrosine) could predict incident diabetes 12 years prior to the
diagnosis with a more than 5-fold higher risk for individuals
in top quartile. More recently, the predictive value of BCAAs
has been validated in two large cohorts [29,30]. Besides,
baseline BCAA concentrations were positively associated
with IR measured 18 months later in healthy children and
adolescents [31].
Finally, there is evidence from interventional studies that
supports the relationship between BCAAs and IR. In
participants from a weight loss trial, Shah et al. [32]
demonstrated that the baseline BCAAs and related catabolites
were able to predict the improvement of IR, independent of
the amount of weight loss. Although it is well known that
gastric bypass surgery (GBP) has more benecial effects on
the glycemic control than dietary intervention in patients with
morbid obesity and T2DM when assuming an equivalent
weight loss, the mechanisms underlying this phenomenon
remain elusive. Comparing the circulating amino acids and
acylcarnitine proles between subjects receiving GBP or
dietary intervention, Lafferere et al. [33] found that BCAAs
were signicantly decreased after GBP but not after dietary
intervention [33], and BCAAs uniquely correlated with IR,
suggesting that the better improvement of glucose home-
ostasis after GBP could in part be attributed to the changes in
BCAAs [28]. Taken together, these studies clearly unveil a
54 Insulin resistance and branched-chain amino acids
BCAA-IR relationship.
As many clinical and experimental measures such as
obesity, blood lipids and inammatory cytokines have been
conrmed to be risk factors for insulin resistance, it is natural
to ask whether BCAAs are related to these risk factors.
However, few studies have addressed this issue to date. Apart
form the long-recognized linkage between BCAAs and
obesity, Floegel et al. [29] observed that a metabolite factor
(in PCA analysis) comprising BCAAs, aromatic amino acids,
propionylcarnitine, hexose and diacyl-phosphatidylcholines
was positively correlated with triglycerides and liver
enzymes. On the other hand, there is emerging evidence
that BCAAs associate with insulin resistance and T2DM
beyond conventional risk factors. For example, Wang-Sattler
et al. [30] reported that BCAAs were predictive for incident
T2DM even after adjustment for age, sex, BMI, physical
activity, alcohol intake, smoking, systolic blood pressure and
HDL cholesterol, suggesting that BCAAs may exert unique
effects on insulin resistance and T2DM risk.
Why are BCAAs elevated in IR condition?
Since BCAAs are essential amino acids, they cannot be
synthesized de novo in organisms. Therefore, dietary protein
can have a signicantly impact on BCAAs in humans.
However, the elevation of BCAAs could occur in obesity
even after overnight fast [14,15]. In addition, there was no
dietary difference between insulin-resistant and-sensitive
individuals in either Asia-Indian or Chinese, as investigated
by Tai et al. [27]. More specically, Wang et al. [28] reported
comparable intakes of valine, leucine and isoleucine among
the study groups of the Framingham cohort, indicating that
increased protein consumption is unlikely to be the major
driving force behind the abnormally high levels of BCAAs
observed in the overweight/obese or insulin resistant subjects.
Another potential explanation for the rise in BCAAs could be
the accelerated protein degradation, which has been reported
in both humans [34] and animal models [35,36] of IR.
Recently, an increasing body of evidence suggests that
impaired BCAA catabolism, especially in adipose tissue,
contributes to the rise in BCAAs in obesity and insulin
resistant states. In two rodent models of obesity (ob/ob mice
and Zucker rats), reduced expression of mitochondrial BCAT
and the BCKDH complex, which catalyzes the rst two
enzymatic steps of BCAA catabolism [37], was observed in
the adipose tissue of obese mice/rats rather than in lean ones,
concordant with signicant increase in plasma BCAAs [38].
The authors further examined the expression of BCAT and
BCKDH complex in human subjects receiving surgical
weight loss intervention (Roux-en-Y gastric bypass) [39].
Signicantly decreased BCAAs after surgery together with
elevated expression of BCAT and BCKDH complex in
omental and subcutaneous fat were found. Pietiainen et al.
[40] performed global gene expression analysis of 14 pairs of
monozygotic twins discordant for BMI, nding that BCAA
catabolism pathway in adipose tissue was downregulated
signicantly in obese twins and correlated closely with IR.
Via the genetic manipulation of mice, Herman et al. [41]
unexpectedly observed that adipose-specic overexpression
of GLUT4, an important glucose transporter, resulted in the
downregulation of BCAA metabolizing enzymes in the
adipose tissue. Interestingly, circulating BCAAs were
upregulated accordingly in their report, further supporting
an important role of adipose tissue in the modulation of
BCAA homeostasis. In a study of Zucker rats treated with one
of the four peroxisome proliferator-activated receptor
(PPAR) ligands including thiazolidinedione (TZD), BCAA
catabolic pathway in the adipose tissue signicantly corre-
lated with ligand treatment-specic insulin-sensitizing
potency [42]. Treating human subjects with TZDs produced
similar results [43]. While most aberrations of BCAA
metabolism in obesity and insulin resistant states were
found in adipose tissue, Lefort et al. [44] reported lower
BCAA metabolizing enzymes (methyl-malonate-semialde-
hyde dehydrogenase and propionyl-CoA carboxylase ) in
mitochondria isolated from skeletal muscle of the obese
group. However, blood BCAA levels were not measured,
precluding the possibility to evaluate the role of skeletal
muscle in the modulation of circulating BCAAs. These
studies suggest that the decreased catabolism of BCAA in
adipose tissue and possibly skeletal muscle may in part be
responsible for the higher levels of BCAAs in insulin resistant
states including obesity.
The possible mechanisms underlying
altered BCAAs in IR
The mTOR is a serine/threonine kinase that belongs to the
phosphatidylinositol (PI) kinase-related protein kinase family
[45]. The mTOR protein is mainly known for its role in
regulating cell growth, notably via protein synthesis. How-
ever, during recent years, mTOR has been considered as the
central signaling molecule mediating the crosstalk between
amino acids and insulin. Stimulation of mTOR activates p70
ribosomal S6 kinase (p70S6K), a key mediator of the protein
synthesis cascade [46] and subsequently leads to the
phosphorylation of its downstream target, ribosomal protein
S6 kinase (S6K). This results in the translation of mRNAs
which encode for ribosomes and transcription factors.
Moreover, activation of S6K1 leads to serine/threonine
phosphorylation and therefore the inhibition of insulin
receptor substrate (IRS)-1, not only interfering with the
normal insulin signaling, but also causing the proteosomal
degradation of IRS-1 [4750].
Leucine stimulates IRS-1 Ser-636/639 phosphorylation in
a rapamycin-sensitive manner in both 3T3-L1 adipocytes and
L6 myotubes in vitro [51]. BCAA supplementation was
shown to increase the activation of mTOR and subsequent
Jingyi Lu et al. 55
S6K1 phosphorylation, coupled with IRS-1 Ser-307 phos-
phorylation in the skeletal muscle of rats [17]. Likewise,
Tremblay et al. [52] reported increased phosphorylation of
the IRS-1 Ser-1011 via mTOR/p70S6K/S6K1 pathway and
decreased insulin-induced phosphoinositide 3-kinase (PI3K)
activity, which is critical for insulin signaling, in human
skeletal muscle under amino acid infusion.
In addition to the well-established mTOR/p70S6K/S6K1
pathway, it has been shown that leucine deprivation
signicantly increased the activation of MAP-activated
protein kinase (AMPK) both in the livers of mice and in
HepG2 cells as compared with controls [53]. The down-
regulation of AMPK decreased insulin-stimulated phosphor-
ylation of insulin receptor and protein kinase B (AKT),
indicative of impaired insulin signaling. General control
nonderepressible (GCN) 2 is a sensor of amino acid
deprivation that triggers a repression of global protein
synthesis while simultaneously inducing translation of
specic proteins [54,55]. In the same study mention above,
Xiao et al. [53] revealed that leucine deprivation was
associated with increased phosphorylation of GCN2 and
concurrent improvement of insulin sensitivity. Intriguingly,
GCN2 acted as an upstream inhibitor of mTOR as evidenced
by the inability of leucine deprivation to blunt mTOR
activation in Gcn2
/
mice. Moreover, Guo et al. [56] found
that the expression of SREBP-1c, a key transcriptional
activator of the de novo lipogenic pathway, was signicantly
downregulated in a GCN2-dependent manner in livers of
mice fed a leucine-decient diet, accompanied by diminished
levels of several lipogenic enzymes and a dramatic loss of
abdominal adipose mass. This phenomenon implies that,
insulin signaling aside, BCAAs could impact on lipid
metabolism and subsequently exert an indirect effect on
insulin sensitivity.
It needs to be pointed out that an extensive review of
previous studies presents a more complex picture regarding
the role of BCAAs in the modulation of insulin sensitivity.
For instance, Macotela et al. [57] reported that doubling
dietary leucine in mice under high fat diet (HFD) caused
marked improvement in glucose tolerance, decreased hepatic
steatosis and decreased inammation in adipose tissue despite
the enhanced activation of mTOR/p70S6K pathway, suggest-
ing that leucine may modify multiple signaling pathways in
multiple tissue, the precise mechanisms of which remain
largely unknown.
Conclusions and future perspectives
The research on BCAAs and IR presents a general picture of
the BCAAs-IR relationship (Fig. 1): (1) the expression of
genes related to BCAA catabolism was downregulated in
adipose tissue and possibly skeletal muscle, leading to BCAA
accumulations, although the mechanisms were not fully
Fig. 1 A schematic view of the contributions of BCAAs to development of insulin resistance. IR, insulin receptor.
56 Insulin resistance and branched-chain amino acids
understood; (2) The elevated BCAAs could then activate the
mTOR/p70S6K, AMPK and GCN2 pathways, thereby
contributing to the development of IR.
However, it is noteworthy that some conicting results
have been reported concerning the role of BCAAs in the
regulation of IR. For instance, using a genetic mouse model,
She et al. [38] reported that the elevation of BCAAs was
accompanied with increased energy expenditure and better
insulin sensitivity. Nishitani et al. [9] reported that leucine
and isoleucine improved glucose metabolism by promoting
glucose uptake in skeletal muscle in rats with liver cirrhosis.
Leucine supplementation was shown to signicantly improve
glucose intolerance and insulin resistance caused by HFD
[57] and similarly, leucine supplementation via drinking
water prevented HFD-induced obesity and hyperglycemia
[58], while no effect of leucine supplementation on the HFD-
induced obesity was observed by Nairizi et al. [59]. Given
that most studies favoring a benecial effect of BCAAs on IR
were based on dietary supplementation, one possible
explanation for the discrepancies could be that BCAAs are
not direct contributors to IR and that its elevation could
simply be a marker of the biological perturbations driving the
development of IR, which merits further investigation. It is
also interesting to note that these studies were conducted in
animal models while recent population-based human studies
with large sample size indicated that the increase of BCAAs is
an early event of T2DM, raising the possibility that the effects
of BCAAs on IR may be species-specic.
In conclusion, recent studies suggest a close relationship
between BCAAs and IR and demonstrate that BCAAs may
play a major role in the modulation of insulin action, despite
some inconsistent results reported from different laboratories.
Progress in this eld will denitely deepen our understanding
in insulin resistant states such as obesity, metabolic syndrome
and T2DM. In addition, BCAA metabolic pathways may also
serve as potential targets for the treatment of metabolic
disorders.
Acknowledgements
This work was supported by grants fromthe National Basic Research
Program of China (973 Program, 2011CB504001) and the National
Natural Science Foundation of China (Grant Nos. 81100590 and
81170760).
References
1. Lewis GF, Carpentier A, Adeli K, Giacca A. Disordered fat storage
and mobilization in the pathogenesis of insulin resistance and type 2
diabetes. Endocr Rev 2002; 23(2): 201229
2. Grundy SM, Brewer HB Jr, Cleeman JI, Smith SC Jr, Lenfant C.
Denition of metabolic syndrome: report of the National Heart,
Lung, and Blood Institute/American Heart Association conference
on scientic issues related to denition. Arterioscler Thromb Vasc
Biol 2004; 24(2): e13e18
3. Rader DJ. Effect of insulin resistance, dyslipidemia, and intra-
abdominal adiposity on the development of cardiovascular disease
and diabetes mellitus. Am J Med 2007; 120(3 Suppl 1): S12S18
4. World Health Organization. Obesity and overweight: Fact sheet N
311. 2012
5. American Diabetes Association. Diagnosis and classication of
diabetes mellitus. Diabetes Care 2009; 32(Suppl 1): S62S67
6. Layman DK. The role of leucine in weight loss diets and glucose
homeostasis. J Nutr 2003; 133(1): 261S267S
7. Doi M, Yamaoka I, Nakayama M, Mochizuki S, Sugahara K,
Yoshizawa F. Isoleucine, a blood glucose-lowering amino acid,
increases glucose uptake in rat skeletal muscle in the absence of
increases in AMP-activated protein kinase activity. J Nutr 2005; 135
(9): 21032108
8. Doi M, Yamaoka I, Nakayama M, Sugahara K, Yoshizawa F.
Hypoglycemic effect of isoleucine involves increased muscle
glucose uptake and whole body glucose oxidation and decreased
hepatic gluconeogenesis. Am J Physiol Endocrinol Metab 2007; 292
(6): E1683E1693
9. Nishitani S, Takehana K, Fujitani S, Sonaka I. Branched-chain
amino acids improve glucose metabolism in rats with liver cirrhosis.
Am J Physiol Gastrointest Liver Physiol 2005; 288(6): G1292
G1300
10. Cota D, Proulx K, Smith KA, Kozma SC, Thomas G, Woods SC,
Seeley RJ. Hypothalamic mTOR signaling regulates food intake.
Science 2006; 312(5775): 927930
11. Baum JI, Layman DK, Freund GG, Rahn KA, Nakamura MT,
Yudell BE. A reduced carbohydrate, increased protein diet stabilizes
glycemic control and minimizes adipose tissue glucose disposal in
rats. J Nutr 2006; 136(7): 18551861
12. Layman DK, Walker DA. Potential importance of leucine in
treatment of obesity and the metabolic syndrome. J Nutr 2006; 136
(1 Suppl): 319S323S
13. Caballero B, Finer N, Wurtman RJ. Plasma amino acids and insulin
levels in obesity: response to carbohydrate intake and tryptophan
supplements. Metabolism 1988; 37(7): 672676
14. Felig P, Marliss E, Cahill GF Jr. Plasma amino acid levels and
insulin secretion in obesity. N Engl J Med 1969; 281(15): 811816
15. Felig P, Marliss E, Cahill GF Jr. Are plasma amino acid levels
elevated in obesity? N Engl J Med 1970; 282(3): 166
16. Marchesini G, Bianchi G, Rossi B, Muggeo M, Bonora E. Effects of
hyperglycaemia and hyperinsulinaemia on plasma amino acid levels
in obese subjects with normal glucose tolerance. Int J Obes Relat
Metab Disord 2000; 24(5): 552558
17. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF,
Haqq AM, Shah SH, Arlotto M, Slentz CA, Rochon J, Gallup D,
Ilkayeva O, Wenner BR, Yancy WS Jr, Eisenson H, Musante G,
Surwit RS, Millington DS, Butler MD, Svetkey LP. A branched-
chain amino acid-related metabolic signature that differentiates
obese and lean humans and contributes to insulin resistance. Cell
Metab 2009; 9(4): 311326
18. Shimomura Y, Honda T, Shiraki M, Murakami T, Sato J, Kobayashi
H, Mawatari K, Obayashi M, Harris RA. Branched-chain amino
acid catabolism in exercise and liver disease. J Nutr 2006; 136(1
Suppl): 250S253S
19. Shimomura Y, Obayashi M, Murakami T, Harris RA. Regulation of
Jingyi Lu et al. 57
branched-chain amino acid catabolism: nutritional and hormonal
regulation of activity and expression of the branched-chain alpha-
keto acid dehydrogenase kinase. Curr Opin Clin Nutr Metab Care
2001; 4(5): 419423
20. Sweatt AJ, Wood M, Suryawan A, Wallin R, Willingham MC,
Hutson SM. Branched-chain amino acid catabolism: unique
segregation of pathway enzymes in organ systems and peripheral
nerves. Am J Physiol Endocrinol Metab 2004; 286(1): E64E76
21. Wei J, Xie G, Ge S, Qiu Y, Liu W, Lu A, Chen T, Li H, Zhou Z, Jia
W. Metabolic transformation of DMBA-induced carcinogenesis and
inhibitory effect of salvianolic acid b and breviscapine treatment. J
Proteome Res 2012; 11(2): 13021316
22. Harris RA, Hawes JW, Popov KM, Zhao Y, Shimomura Y, Sato J,
Jaskiewicz J, Hurley TD. Studies on the regulation of the
mitochondrial alpha-ketoacid dehydrogenase complexes and their
kinases. Adv Enzyme Regul 1997; 37: 271293
23. Popov KM, Zhao Y, Shimomura Y, Kuntz MJ, Harris RA.
Branched-chain alpha-ketoacid dehydrogenase kinase. Molecular
cloning, expression, and sequence similarity with histidine protein
kinases. J Biol Chem 1992; 267(19): 1312713130
24. Damuni Z, Reed LJ. Purication and properties of the catalytic
subunit of the branched-chain alpha-keto acid dehydrogenase
phosphatase from bovine kidney mitochondria. J Biol Chem 1987;
262(11): 51295132
25. Huffman KM, Shah SH, Stevens RD, Bain JR, Muehlbauer M,
Slentz CA, Tanner CJ, Kuchibhatla M, Houmard JA, Newgard CB,
Kraus WE. Relationships between circulating metabolic intermedi-
ates and insulin action in overweight to obese, inactive men and
women. Diabetes Care 2009; 32(9): 16781683
26. Shaham O, Wei R, Wang TJ, Ricciardi C, Lewis GD, Vasan RS, Carr
SA, Thadhani R, Gerszten RE, Mootha VK. Metabolic proling of
the human response to a glucose challenge reveals distinct axes of
insulin sensitivity. Mol Syst Biol 2008; 4: 214
27. Tai ES, Tan ML, Stevens RD, Low YL, Muehlbauer MJ, Goh DL,
Ilkayeva OR, Wenner BR, Bain JR, Lee JJ, Lim SC, Khoo CM,
Shah SH, Newgard CB. Insulin resistance is associated with a
metabolic prole of altered protein metabolism in Chinese and
Asian-Indian men. Diabetologia 2010; 53(4): 757767
28. Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E,
Lewis GD, Fox CS, Jacques PF, Fernandez C, ODonnell CJ, Carr
SA, Mootha VK, Florez JC, Souza A, Melander O, Clish CB,
Gerszten RE. Metabolite proles and the risk of developing
diabetes. Nat Med 2011; 17(4): 448453
29. Floegel A, Stefan N, Yu Z, Muhlenbruch K, Drogan D, Joost HG,
Fritsche A, Haring HU, Hrabe de Angelis M, Peters A, Roden M,
Prehn C, Wang-Sattler R, Illig T, Schulze MB, Adamski J, Boeing
H, Pischon T. Identication of Serum Metabolites Associated With
Risk of Type 2 Diabetes Using a Targeted Metabolomic Approach.
Diabetes 2012 Oct 4. [Epub ahead of print] doi: 10.2337/db12-0495
30. Wang-Sattler R, Yu Z, Herder C, Messias AC, Floegel A, He Y,
Heim K, Campillos M, Holzapfel C, Thorand B, Grallert H, Xu T,
Bader E, Huth C, Mittelstrass K, Dring A, Meisinger C, Gieger C,
Prehn C, Roemisch-Margl W, Carstensen M, Xie L, Yamanaka-
Okumura H, Xing G, Ceglarek U, Thiery J, Giani G, Lickert H, Lin
X, Li Y, Boeing H, Joost HG, de Angelis MH, Rathmann W, Suhre
K, Prokisch H, Peters A, Meitinger T, Roden M, Wichmann HE,
Pischon T, Adamski J, Illig T. Novel biomarkers for pre-diabetes
identied by metabolomics. Mol Syst Biol 2012; 8: 615
31. McCormack SE, Shaham O, McCarthy MA, Deik AA, Wang TJ,
Gerszten RE, Clish CB, Mootha VK, Grinspoon SK, Fleischman A.
Circulating branched-chain amino acid concentrations are asso-
ciated with obesity and future insulin resistance in children and
adolescents. Pediatr Obes 2013; 8(1): 5261
32. Shah SH, Crosslin DR, Haynes CS, Nelson S, Turer CB, Stevens
RD, Muehlbauer MJ, Wenner BR, Bain JR, Laferrre B,
Gorroochurn P, Teixeira J, Brantley PJ, Stevens VJ, Hollis JF,
Appel LJ, Lien LF, Batch B, Newgard CB, Svetkey LP. Branched-
chain amino acid levels are associated with improvement in insulin
resistance with weight loss. Diabetologia 2012; 55(2): 321330
33. Laferrre B, Reilly D, Arias S, Swerdlow N, Gorroochurn P, Bawa
B, Bose M, Teixeira J, Stevens RD, Wenner BR, Bain JR,
Muehlbauer MJ, Haqq A, Lien L, Shah SH, Svetkey LP, Newgard
CB. Differential metabolic impact of gastric bypass surgery versus
dietary intervention in obese diabetic subjects despite identical
weight loss. Sci Transl Med 2011; 3(80):80re2
34. Luzi L, Castellino P, DeFronzo RA. Insulin and hyperaminoacide-
mia regulate by a different mechanism leucine turnover and
oxidation in obesity. Am J Physiol 1996; 270(2 Pt 1): E273E281
35. Argils JM, Busquets S, Alvarez B, Lpez-Soriano FJ. Mechanism
for the increased skeletal muscle protein degradation in the obese
Zucker rat. J Nutr Biochem 1999; 10(4): 244248
36. Wang X, Hu Z, Hu J, Du J, Mitch WE. Insulin resistance accelerates
muscle protein degradation: Activation of the ubiquitin-proteasome
pathway by defects in muscle cell signaling. Endocrinology 2006;
147(9): 41604168
37. Suryawan A, Hawes JW, Harris RA, Shimomura Y, Jenkins AE,
Hutson SM. A molecular model of human branched-chain amino
acid metabolism. Am J Clin Nutr 1998; 68(1): 7281
38. She P, Reid TM, Bronson SK, Vary TC, Hajnal A, Lynch CJ,
Hutson SM. Disruption of BCATmin mice leads to increased energy
expenditure associated with the activation of a futile protein
turnover cycle. Cell Metab 2007; 6(3): 181194
39. She P, Van Horn C, Reid T, Hutson SM, Cooney RN, Lynch CJ.
Obesity-related elevations in plasma leucine are associated with
alterations in enzymes involved in branched-chain amino acid
metabolism. Am J Physiol Endocrinol Metab 2007; 293(6): E1552
E1563
40. Pietilinen KH, Naukkarinen J, Rissanen A, Saharinen J, Ellonen P,
Kernen H, Suomalainen A, Gtz A, Suortti T, Yki-Jrvinen H,
Oresic M, Kaprio J, Peltonen L. Global transcript proles of fat in
monozygotic twins discordant for BMI: pathways behind acquired
obesity. PLoS Med 2008; 5(3): e51
41. Herman MA, She P, Peroni OD, Lynch CJ, Kahn BB. Adipose
tissue branched chain amino acid (BCAA) metabolism modulates
circulating BCAA levels. J Biol Chem 2010; 285(15): 1134811356
42. Hsiao G, Chapman J, Ofrecio JM, Wilkes J, Resnik JL, Thapar D,
Subramaniam S, Sears DD. Multi-tissue, selective PPAR modula-
tion of insulin sensitivity and metabolic pathways in obese rats. Am
J Physiol Endocrinol Metab 2011; 300(1): E164E174
43. Sears DD, Hsiao G, Hsiao A, Yu JG, Courtney CH, Ofrecio JM,
Chapman J, Subramaniam S. Mechanisms of human insulin
resistance and thiazolidinedione-mediated insulin sensitization.
Proc Natl Acad Sci USA 2009; 106(44): 1874518750
44. Lefort N, Glancy B, Bowen B, Willis WT, Bailowitz Z, De Filippis
58 Insulin resistance and branched-chain amino acids
EA, Brophy C, Meyer C, Hjlund K, Yi Z, Mandarino LJ. Increased
reactive oxygen species production and lower abundance of
complex I subunits and carnitine palmitoyltransferase 1B protein
despite normal mitochondrial respiration in insulin-resistant human
skeletal muscle. Diabetes 2010; 59(10): 24442452
45. Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes
Dev 2004; 18(16): 19261945
46. Hara K, Maruki Y, Long X, Yoshino K, Oshiro N, Hidayat S,
Tokunaga C, Avruch J, Yonezawa K. Raptor, a binding partner of
target of rapamycin (TOR), mediates TORaction. Cell 2002; 110(2):
177189
47. Haruta T, Uno T, Kawahara J, Takano A, Egawa K, Sharma PM,
Olefsky JM, Kobayashi M. A rapamycin-sensitive pathway down-
regulates insulin signaling via phosphorylation and proteasomal
degradation of insulin receptor substrate-1. Mol Endocrinol 2000;
14(6): 783794
48. OConnor JC, Freund GG. Vanadate and rapamycin synergistically
enhance insulin-stimulated glucose uptake. Metabolism 2003; 52
(6): 666674
49. Pederson TM, Kramer DL, Rondinone CM. Serine/threonine
phosphorylation of IRS-1 triggers its degradation: possible regula-
tion by tyrosine phosphorylation. Diabetes 2001; 50(1): 2431
50. Sun XJ, Rothenberg P, Kahn CR, Backer JM, Araki E, Wilden PA,
Cahill DA, Goldstein BJ, White MF. Structure of the insulin
receptor substrate IRS-1 denes a unique signal transduction
protein. Nature 1991; 352(6330): 7377
51. Tzatsos A, Kandror KV. Nutrients suppress phosphatidylinositol 3-
kinase/Akt signaling via raptor-dependent mTOR-mediated insulin
receptor substrate 1 phosphorylation. Mol Cell Biol 2006; 26(1):
6376
52. Tremblay F, Krebs M, Dombrowski L, Brehm A, Bernroider E,
Roth E, Nowotny P, Waldhusl W, Marette A, Roden M.
Overactivation of S6 kinase 1 as a cause of human insulin resistance
during increased amino acid availability. Diabetes 2005; 54(9):
26742684
53. Xiao F, Huang Z, Li H, Yu J, Wang C, Chen S, Meng Q, Cheng Y,
Gao X, Li J, Liu Y, Guo F. Leucine deprivation increases hepatic
insulin sensitivity via GCN2/mTOR/S6K1 and AMPK pathways.
Diabetes 2011; 60(3): 746756
54. Bruhat A, Jousse C, Fafournoux P. Amino acid limitation regulates
gene expression. Proc Nutr Soc 1999; 58(3): 625632
55. Kilberg MS, Pan YX, Chen H, Leung-Pineda V. Nutritional control
of gene expression: how mammalian cells respond to amino acid
limitation. Annu Rev Nutr 2005; 25(1): 5985
56. Guo F, Cavener DR. The GCN2 eIF2alpha kinase regulates fatty-
acid homeostasis in the liver during deprivation of an essential
amino acid. Cell Metab 2007; 5(2): 103114
57. Macotela Y, Emanuelli B, Bng AM, Espinoza DO, Boucher J,
Beebe K, Gall W, Kahn CR. Dietary leucinean environmental
modier of insulin resistance acting on multiple levels of
metabolism. PLoS ONE 2011; 6(6): e21187
58. Zhang Y, Guo K, LeBlanc RE, Loh D, Schwartz GJ, Yu YH.
Increasing dietary leucine intake reduces diet-induced obesity and
improves glucose and cholesterol metabolism in mice via multi-
mechanisms. Diabetes 2007; 56(6): 16471654
59. Nairizi A, She P, Vary TC, Lynch CJ. Leucine supplementation of
drinking water does not alter susceptibility to diet-induced obesity in
mice. J Nutr 2009; 139(4): 715719
Jingyi Lu et al. 59

Você também pode gostar