Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3386 2023-12-13 16:24:44 |
2 Reference format revised. Meta information modification 3386 2023-12-19 03:52:21 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Yang, W.; Jiang, W.; Guo, S. Regulation of Macronutrients during Type 2 Diabetes Mellitus. Encyclopedia. Available online: https://encyclopedia.pub/entry/52699 (accessed on 08 July 2024).
Yang W, Jiang W, Guo S. Regulation of Macronutrients during Type 2 Diabetes Mellitus. Encyclopedia. Available at: https://encyclopedia.pub/entry/52699. Accessed July 08, 2024.
Yang, Wanbao, Wen Jiang, Shaodong Guo. "Regulation of Macronutrients during Type 2 Diabetes Mellitus" Encyclopedia, https://encyclopedia.pub/entry/52699 (accessed July 08, 2024).
Yang, W., Jiang, W., & Guo, S. (2023, December 13). Regulation of Macronutrients during Type 2 Diabetes Mellitus. In Encyclopedia. https://encyclopedia.pub/entry/52699
Yang, Wanbao, et al. "Regulation of Macronutrients during Type 2 Diabetes Mellitus." Encyclopedia. Web. 13 December, 2023.
Regulation of Macronutrients during Type 2 Diabetes Mellitus
Edit

Insulin resistance is an important feature of metabolic syndrome and a precursor of type 2 diabetes mellitus (T2DM). Overnutrition-induced obesity is a major risk factor for the development of insulin resistance and T2DM. The intake of macronutrients plays a key role in maintaining energy balance. The components of macronutrients distinctly regulate insulin sensitivity and glucose homeostasis. Precisely adjusting the beneficial food compound intake is important for the prevention of insulin resistance and T2DM. 

macronutrients insulin resistance glucose homeostasis type 2 diabetes mellitus

1. Introduction

Type 2 diabetes mellitus (T2DM) has become a global health issue that is tightly correlated with the prevalence of obesity and other chronic diseases. T2DM is caused by systemic insulin resistance and impaired insulin secretion in pancreatic β-cells, leading to disorders of carbohydrate, protein, and lipid metabolism [1]. Insulin resistance is a hallmark of prediabetes and gradually contributes to the development of T2DM. Insulin resistance refers to an impaired ability of insulin to lower blood glucose in target tissues at a normal plasma insulin level. Pancreatic β-cells secrete excessive insulin to compensate for the outcome of insulin resistance. Thus, fasting plasma insulin levels rise, and hyperinsulinemia eventually develops [2]. Although it is controversial about the primary defect between insulin resistance and hyperinsulinemia, they together culminate in eventual β-cell failure, resulting in hyperglycemia [3][4].
Insulin receptor (IR) is a tyrosine kinase receptor that is activated upon insulin binding, recruiting downstream substrates such as insulin receptor substrate (IRS). This initiation sets off the proximal insulin signaling pathway. Phosphorylated IRS, in turn, triggers the phosphoinositide 3-kinase (PI3K) → protein kinase B (AKT) signaling cascade, regulating the activity of critical distal downstream targets, including glucose transporter type 4 (GLUT4), mammalian target of rapamycin complex 1 (mTORC1), and forkhead box protein O1 (FoxO1), regulating glucose and energy homeostasis [5][6]. Insulin resistance can manifest at multiple cellular levels, including the desensitization of the insulin receptor at the cell surface, inhibition of IRS function via protein degradation, suppression of PI3K activity, an inability to inhibit FoxO1-induced transcriptional changes, and reduced insulin clearance from the bloodstream [6][7].
Metabolic organs cooperatively respond to nutrient intake and maintain energy balance. The interaction of nutrients, especially macronutrients, with the gastrointestinal tract stimulates the release of incretin hormones such as glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) [8]. GLP-1 improves insulin sensitivity in peripheral tissues through increasing insulin secretion, attenuating inflammation response and endoplasmic reticulum (ER) stress, increasing GLUT-4 expression, and enhancing insulin signal transduction [9][10][11][12]. GIP stimulates the release of both insulin and glucagon in the pancreas. Therefore, incretin hormones play a key role in regulating insulin sensitivity and glucose homeostasis. Nutrient ingestion, especially carbohydrates, increases blood glucose and stimulates insulin secretion in the pancreas, thereby suppressing glucose production in the liver, increasing lipogenesis in the adipose tissue, promoting glucose uptake in the skeletal muscle, and regulating glucose homeostasis. Overnutrition-induced obesity is highly related to insulin resistance, primarily through multiple pathological mechanisms. The nutrient-induced GLP-1 secretion is significantly reduced in obese individuals [13][14], which may contribute to systemic insulin resistance. During overnutrition, toxic metabolites including ceramides, diacylglycerol (DAG), and nonesterified fatty acids (NEFA) accumulate and stimulate the activity of protein kinase C (PKC), leading to the Ser/Thr phosphorylation of IR and IRS, thereby impairing insulin sensitivity [2]. Overnutrition elevates branched-chain amino acid levels in the bloodstream, activating mTORC1 and inhibiting IRS function [15]. Proinflammatory cytokine levels in circulation are increased during obesity, which induces Ser/Thr phosphorylation of IRS and inhibits insulin signaling by activating JNK and IKKβ [16][17][18]. Hyperinsulinemia associated with obesity leads to insulin resistance by inhibiting the activity of IRS [19].

2. Insulin and Glucagon Action and Insulin Resistance

2.1. Molecular Basis of Insulin and Glucagon Signaling

Insulin and glucagon are pivotal hormones that play essential roles in regulating glucose homeostasis. These two hormones cooperate tightly to maintain blood glucose levels within a narrow range, preventing both hyperglycemia and hypoglycemia under certain health conditions. The glucagon-to-insulin ratio is strongly linked to hyperglycemia in patients with type 2 diabetes [20].
Insulin, a peptide hormone secreted from pancreatic β-cells, plays a critical role in orchestrating the anabolic response to nutrient intake, especially the intake of glucose, fatty acids, and amino acids [21]. Insulin regulates blood glucose balance by increasing glucose uptake in skeletal muscle and fat tissues while suppressing hepatic glucose production (HGP). Insulin actions are induced by the intrinsic tyrosine kinase activity of the insulin receptor (IR). Insulin triggers conformational changes and autophosphorylation of IR, leading to the recruitment and phosphorylation of IRS and SH3-containing protein (Shc). IRS activates the PI3K → AKT pathway to govern insulin’s metabolic functions, while Shc activates the Ras → MAPK pathway, which mediates growth and differentiation at cellular and organismal levels [22]. Additionally, IR also exerts control over apoptosis, senescence, and the cell cycle, independent of ligand and tyrosine kinase activity [23]. In skeletal muscle and adipose tissues, insulin promotes the translocation of GLUT4 from the cytoplasm to the cell membrane, thereby promoting glucose uptake [24]. Insulin-stimulated GLUT4 trafficking is mediated by the PI3K → AKT → AS160 signaling cascade [25][26][27]. AS160, a Rab GTPase-activating protein, maintains Rab proteins in an inactive GDP form and results in GLUT4 retention in the cytoplasm. Phosphorylation of AS160 by AKT inactivates its Rab GTPase activity, resulting in an increase in the active GTP form of Rab, thereby promoting GLUT4 trafficking [24][25]. In addition, AKT-induced PIKfyve phosphorylation plays a role in regulating insulin-stimulated GLUT4 trafficking, potentially through PtdIns 3,5-P2 [28][29][30]. In the liver, insulin inhibits glycogenolysis and gluconeogenesis, thus suppressing glucose release. Insulin activates glycogen synthase by the PI3K → AKT → GSK-3 signaling pathway. AKT stimulates phosphorylation of GSK-3 and inhibits its kinase activity, thereby dephosphorylating glycogen synthase, stimulating its activity, and promoting glycogen accumulation [31]. The IRS → PI3K → AKT → FoxO1 signaling pathway in the liver is primarily responsible for insulin action on glucose homeostasis [32]. Insulin stimulates phosphorylation of FoxO1 at S253 via the activation of AKT, thereby decreasing FoxO1-induced expression of genes responsible for gluconeogenesis such as phosphoenolpyruvate carboxykinase 1 (Pck1) and glucose-6-phosphatase (G6pase) and suppressing HGP [33][34][35]. The central nervous system (CNS), such as the hypothalamus, plays a pivotal role in regulating systemic insulin sensitivity and glucose homeostasis [36]. The activation of insulin signaling in agouti-related peptide (AgRP) but not pro-opiomelanocortin (POMC) neurons suppresses HGP through the hepatic vagal nerve [37]. Insulin induces the hyperpolarization of AgRP neurons and decreases their activity, thus leading to the activation of IL-6-STAT3 signaling and downregulating hepatic gluconeogenic genes, including Pck1 and G6pase [37][38].
Glucagon is secreted by the pancreatic α-cells, acting as a catabolic hormone and regulating glucose homeostasis. Glucagon promotes HGP through stimulation of glycogenolysis and gluconeogenesis, thereby maintaining euglycemia under fasting conditions. The action of glucagon is mediated by the glucagon receptor (Gcgr), a G protein-coupled receptor [39]. Upon glucagon binding, Gcgr activates Gs protein, thereby stimulating adenylate cyclase, elevating cyclic adenosine monophosphorylate (cAMP), and then activating cAMP-dependent protein kinase A (PKA) and exchange protein directly activated by cAMP 2 (EPAC2) [40]. Active PKA, in turn, stimulates the activity of phosphorylase kinase, which then converts glycogen phosphorylase b (PYG b) into the active form PYG a and promotes glycogen breakdown [41]. Glucagon promotes gluconeogenesis through the activation of two key transcriptional factors, cAMP response element-binding protein (CREB) and FoxO1 [42]. Activation of PKA by glucagon induces phosphorylation of CREB at S133, resulting in the formation of the CREB-TORC2 complex and increasing transcription of its downstream gluconeogenic genes, including Ppargc1a, Pck1, and G6pase [43][44]. Glucagon stimulates phosphorylation of FoxO1 at S273 via both PKA and EPAC2 → p38 signaling pathways, enhancing FoxO1 stability and nuclear localization [45][46].

2.2. The Molecular Basis of Insulin Resistance by Targeting FoxO1

The molecular mechanisms of insulin resistance have been extensively reviewed [2][5][6][22]. During obesity, lipotoxicity, chronic inflammation, hyperglycemia, hyperinsulinemia, mitochondrial dysfunction, and ER stress stimulate the activity of Ser/Thr kinase and impair insulin sensitivity by phosphorylating IR, IRS, and AKT proteins, resulting in insulin resistance. Insulin resistance is a critical mechanism underlying various metabolic disorders. Over the past decade, scholars have proven that FoxO1 is one of the key factors that link insulin resistance and metabolic disorders. Studies from many labs, including our own, have demonstrated that FoxO1 promotes HGP by upregulating the expression of gluconeogenic genes, including Pck1 and G6pase [47][48]. Insulin stimulates the activity of AKT and phosphorylates FoxO1 at T24, S253, and S316, thereby suppressing FoxO1 activity and inhibiting HGP [34][35][49][50]. Deletion of liver IRS1 and 2 in mice induces systemic insulin resistance, leading to diabetic symptoms that include hyperglycemia and hyperinsulinemia. Notably, these diabetic symptoms are normalized when hepatic FoxO1 is deficient [32]. The scholars recently uncovered that glucagon stimulates the phosphorylation of FoxO1-S273 through the cAMP → PKA and cAMP → EPAC2 → p38α signaling pathways; this enhances FoxO1 protein stability and promotes its nuclear localization. It also found that FoxO1-S273 phosphorylation prevents insulin-mediated FoxO1 degradation in vitro and impairs glucose tolerance in vivo [45][46]. These findings suggest that FoxO1 is a pivotal mediator connecting glucagon and insulin signaling in the regulation of glucose homeostasis. In addition, FoxO1 plays a crucial role in aging-induced glucose dysregulation and chronic inflammation. It found that the activity of FoxO1 was increased in the livers of old mice. Inhibition of FoxO1 significantly improves glucose homeostasis and attenuates chronic inflammation in Kupffer cells during aging [51]. Activation of FoxO1 impairs hepatic mitochondrial function by controlling heme homeostasis, thereby contributing to insulin resistance-mediated hepatic mitochondrial dysfunction [52][53]. Heme oxygenase-1 (HO-1) is one of the target genes regulated by FoxO1 [52]. HO-1 gain-of-function increases ferrous iron levels and promotes inflammation in livers [54]; these results indicate that the FoxO1 → HO-1 signaling pathway plays a key role in ferrous iron overload and chronic inflammation under insulin-resistant conditions. Activation of FoxO1 increases TGF-β1 levels, impairing glucose and energy metabolism as well as exacerbating CCL4-induced liver fibrosis [55][56]

3. Dietary Carbohydrates and Glucose Homeostasis

3.1. Dietary Sugars and Insulin Resistance

The increased consumption of refined and simple carbohydrates promotes the development of insulin resistance and T2DM. The World Health Organization and the Food and Agriculture Organization recommended a restriction on free sugar intake to prevent T2DM and obesity [57]. The clinical studies show that high fructose intake (>250 g/day) decreases insulin sensitivity and increases adiposity in both healthy and obese individuals [58][59]. However, moderate fructose intake (<100 g/day) has a limited effect on insulin sensitivity and metabolic health [60]. The overconsumption of dietary sugars promotes the development of insulin resistance, both directly and indirectly. Dietary sugar overconsumption promotes a positive energy balance, thereby increasing body weight and fat deposition and indirectly leading to insulin resistance and glucose dysregulation. Additionally, fructose reduces hypothalamic malonyl-CoA levels and increases the hunger hormone ghrelin [61][62][63], thereby stimulating appetite, increasing body weight, and impairing insulin sensitivity. On the other hand, fructose, the sweetest of all naturally occurring carbohydrates, directly causes insulin resistance via increasing hepatic lipid accumulation and promoting inflammation. Fructose is a highly lipogenic sugar and is mainly metabolized in the liver, with very little entering the systemic circulation [64]. Fructose is transported into the liver by GLUT2, phosphorylated into fructose-1-phosphate, and further metabolized to dihydroxyacetone phosphate and glyceraldehyde 3-phosphate, which stimulates de novo lipogenesis [65]. Administration of fructose to mice significantly increases the activity of carbohydrate-responsive element-binding protein (ChREBP) and sterol regulatory element-binding protein 1 (SREBP-1c), two important transcription factors stimulating lipogenesis [66][67].
On the other hand, dietary sugar or fructose impairs immune homeostasis and promotes inflammation, thereby leading to insulin resistance. Dietary fructose is transported into the enterocyte through a specific fructose transporter, GLUT5. Healthy individuals can absorb up to 25 g of fructose [68]. High-fructose diet-induced fructose overload leads to intestinal barrier deterioration and alters gut microbiota composition, thereby increasing endotoxin release, promoting systemic inflammation, and leading to insulin resistance [69][70]. The impairment of the intestinal tissue repair and the gut microbiota environment by fructose leads to intestinal barrier deterioration [71]. Fructose-stimulated endotoxin release increases TNF production in liver macrophages, mediating fructose-induced hepatic insulin resistance and lipogenesis [71][72]. Dietary sugar regulates the crosstalk between the microbiota and intestinal immunity to control insulin resistance and other metabolic disorders. Dietary sugar is a key to disrupting intestinal immune homeostasis via eliminating Th17-inducing microbiota (especially segmented filamentous bacteria).
Low-calorie sweeteners (LCS), including aspartame, saccharin, sucralose, and steviol glycoside, provide an alternative to added sugars and have been used to control body weight gain [73]. The effects of LCS on reduction of caloric intake and body weight gain make LCS a promising approach to controlling blood glucose in patients with T2DM. However, the beneficial effects of LCS are controversial in human studies. In some randomized controlled trials (RCTs), LCSs significantly decreased body weight, body mass index, fat mass, and waist circumference [74]. Contrarily, results from some RCTs fail to show a significant effect of LCSs on weight management [75]

3.2. Dietary Fibers and Insulin Resistance

Dietary fiber is composed of highly complicated substances that include any indigestible carbohydrate and lignin that cannot be degraded in the upper gastrointestinal tract. The viscous, gel-forming, and soluble dietary fiber derived from fruit and certain vegetables suppresses the absorption of macronutrients, reduces postprandial glucose response, and improves lipid profiles. A meta-analysis with 176,117 subjects showed that higher insoluble cereal fiber intake was significantly associated with a lower incidence of diabetes, whereas fruit and vegetable fiber intake had no significant association with the risk of T2DM [76]. Consistently, the consumption of insoluble dietary fiber significantly increases insulin sensitivity in both healthy and diabetic individuals [77][78][79]. However, other studies provide evidence that soluble dietary fibers decrease postprandial blood glucose and increase insulin sensitivity in both nondiabetic and diabetic subjects [80][81]. Compared to soluble dietary fiber, insoluble dietary fiber has a better effect on attenuation of high-fat diet-induced obesity, body fat composition, and insulin resistance in mice [82]. Short-chain fatty acids (SCFAs), including butyrate, propionate, and acetate, are generated by the gut microbiota through fermenting non-digestible dietary fibers and play an important role in the beneficial effects of dietary fibers on insulin resistance and glucose homeostasis. SCFAs stimulate the secretion of the gut hormone anorexigenic peptide YY (PYY) through free fatty acid receptor 2 (FFAR2), thereby increasing satiety, decreasing body weight, and improving insulin sensitivity [83]. Additionally, SCFAs acetate and propionate increase the secretion of GLP-1 via FFAR2 and 3, contributing to the improvement in glucose tolerance and hepatic insulin sensitivity [84][85].

4. Lipid Metabolism and Glucose Homeostasis

4.1. Dietary Fat and Insulin Resistance

Dietary fat is highly associated with the incidence of insulin resistance in both mice and humans. In mice, high-fat diet (60% calories derive from fat) feeding for 13 weeks leads to insulin resistance and glucose intolerance [86]. In humans, a higher total fat intake is significantly associated with increased fasting insulin levels, HbA1c, and 2-h post-load glucose levels [87][88][89]. It is estimated that an increase in dietary fat intake of 40 g/d is correlated with a 3.4-fold increased risk of T2DM [90]. However, several population studies failed to show a significant correlation between total fat intake and the incidence of T2DM, which may be attributed to the quantity and quality of dietary fat. There are mainly two kinds of dietary fats: saturated and unsaturated fats, which play different roles in the development of insulin resistance.

4.2. Molecular Mechanisms of FFA-Induced Insulin Resistance

Lipid overload induces insulin resistance in skeletal muscle and liver, thereby leading to defects in insulin-mediated glucose uptake and suppression of HGP, respectively. The detrimental effect of lipid overload on insulin sensitivity is mainly attributed to the increased circulating free fatty acid (FFA) levels, especially saturated free fatty acids (SFAs) [91]. In the KANWU study, high SFA diet intervention significantly impairs insulin sensitivity in both healthy men and women [92]. Aberrant SFA levels stimulate the inflammatory pathway, increase ROS production, generate insulin resistance-associated lipid production, and impair protein folding homeostasis, thereby leading to insulin resistance.
Long-chain polyunsaturated fatty acids (PUFAs), particularly the n-3 family, play a key role in regulating insulin sensitivity [93]. Several human studies have shown that omega-3 PUFAs improve insulin sensitivity in both obese non-diabetic and diabetic individuals [94][95]. Omega-3 PUFAs improve insulin sensitivity, potentially through anti-inflammation and PPAR activation. Previous studies showed that omega-3 PUFAs significantly attenuated TNF or TLR2/4 agonist-induced inflammation through GPR120 in macrophages [96][97]. Omega-3 PUFAs promote the association of GPR120 and β arrestin-2 and then induce the internalization of the GPR120/β arrestin-2 complex. The cytoplasmic β arrestin-2 interacts with TAB1 and blocks the interaction between TAB1 and TAK1, thereby inhibiting TAK1 activity, downstream IKKβ/NFκB and JNK/AP1 signaling, and improving diet-induced insulin resistance [97]. In addition, the anti-inflammatory role of omega-3 PUFAs is potentially mediated by the small lipid mediators (protectins/resolvins) that are metabolized from omega-3 PUFAs.

5. Protein Metabolism and Glucose Homeostasis

5.1. Dietary Proteins and Insulin Resistance

Dietary protein intake is important for normal growth and development. It is recommended that dietary protein accounts for 10–35% of the total diet. Numerous human studies have reported that a short-term increase in dietary protein consumption significantly decreases body weight. A high-protein diet increases satiety via the gut hormone PYY, stimulates thermogenesis [98], potentially due to the high ATP-consuming protein synthesis, and reduces subsequent energy intake, thereby leading to great weight loss [99][100]. Additionally, protein feeding induces intestinal gluconeogenesis and promotes portal glucose release, which leads to hypothalamic activation through the nerve system around the portal vein and decreases food intake [101][102]. Accordingly, short-term high dietary protein consumption stimulates insulin secretion and reduces blood glucose levels [103]. Consistently, a protein preload significantly decreases postprandial glycemia and increases plasma insulin levels in patients with type 2 diabetes, which is partially attributed to the elevation of GLP-1 and GIP levels [104][105]. A high-protein diet intervention in individuals with type 2 diabetes for 5 weeks improves glucose homeostasis [106]. However, long-term studies (over six months) in humans show that a high-protein diet is significantly associated with increased fasting glucose levels, enhanced gluconeogenesis, and impaired insulin sensitivity [107][108].

5.2. Amino Acid and Insulin Resistance

In obese individuals, most serum amino acid levels are significantly increased, suggesting that amino acids may play a role in the regulation of insulin resistance [109]. A previous study showed that a short-term increase in plasma amino acids induces insulin resistance in skeletal muscle, decreases glycogen synthesis, and reduces whole-body glucose disposal [110][111]. Amino acid infusion stimulates phosphorylation of IRS1 at serine 312 and 636/639 via activation of the mTOR/S6 kinase 1 signaling pathway, thereby decreasing the interaction of the p85 subunit of PI3K with IRS1 and leading to insulin resistance in skeletal muscle [111][112]. In vitro studies in hepatocytes and adipocytes also show that amino acid treatment negatively modulates insulin sensitivity [113][114].
Although an amino acid mixture attenuates insulin sensitivity both in vitro and in vivo, different amino acids have distinct effects on insulin sensitivity. Branched-chain amino acids (BCAAs), including valine, isoleucine, and leucine, are critical nutrient signals in regulating body weight and muscle protein synthesis. In obese patients, plasma BCAA levels are significantly elevated [109]. Previous studies have shown that BCAAs are significantly associated with the risk of insulin resistance in humans, and BCAAs exacerbate diet-induced insulin resistance in mice [115][116][117]. Elevated BCAA levels stimulate the activity of mTOR to hamper insulin signaling via inhibitory phosphorylation of IRS1 or E3 ligase Mul1-mediated AKT2 degradation [109][115]. In addition, abnormal BCAA metabolism during obesity leads to the accumulation of toxic BCAA metabolites, thereby activating stress signaling and promoting insulin resistance [118].

6. Conclusions

T2DM is marked by systemic insulin resistance and β-cell failure-induced insulin deficiency. Overnutrition promotes insulin resistance at multiple levels, including insulin receptor desensitization at the cell surface, inhibition of IRS function through degradation, suppression of PI3K activity, failure to control FoxO1-induced transcriptional changes, and reduced insulin clearance from the bloodstream. Dietary carbohydrate intake induces insulin secretion, and overconsumption of carbohydrates leads to insulin resistance. Fructose is a major driver for the development of insulin resistance through increasing hepatic lipogenesis and impairing gut immunity. Dietary fiber improves insulin sensitivity through gut microbiome-derived SCFAs. Increased dietary fat intake elevates free fatty acid levels, especially unsaturated fatty acids, thereby attenuating insulin sensitivity by inducing pro-inflammatory activity and activating DAG-PKC signaling. PUFA improves insulin resistance through suppression of TLR2/4 signaling and activation of PPAR signaling. The composition of amino acids after dietary protein intake plays a pivotal role in insulin sensitivity and glucose homeostasis. Particularly, increased BCAA levels induce insulin resistance through activation of mTOR-IRS signaling and BCAA metabolite-induced oxidative stress. Short-term protein feeding improves glucose homeostasis, which may be attributed to the increased release of gut hormones, including GLP-1 and GIP. Precision nutrition aims to design a unique nutritional recommendation for each individual according to the combination of an individual’s genetics, metabolome, microbiota, and lifestyle factors, thereby preventing metabolic diseases. Understanding the molecular mechanisms of diet-gene interactions will help us apply precision nutrition to clinical practice by integrating multi-omics approaches.

References

  1. DeFronzo, R.A.; Ferrannini, E.; Groop, L.; Henry, R.R.; Herman, W.H.; Holst, J.J.; Hu, F.B.; Kahn, C.R.; Raz, I.; Shulman, G.I. Type 2 diabetes mellitus. Nat. Rev. Dis. Primers 2015, 1, 15019.
  2. Petersen, M.C.; Shulman, G.I. Mechanisms of insulin action and insulin resistance. Physiol. Rev. 2018, 98, 2133–2223.
  3. Kahn, S. The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 2003, 46, 3–19.
  4. Czech, M.P. Insulin action and resistance in obesity and type 2 diabetes. Nat. Med. 2017, 23, 804–814.
  5. White, M.F.; Kahn, C.R. Insulin action at a molecular level–100 years of progress. Mol. Metab. 2021, 52, 101304.
  6. Guo, S. Insulin signaling, resistance, and the metabolic syndrome: Insights from mouse models to disease mechanisms. J. Endocrinol. 2014, 220, T1.
  7. Najjar, S.M.; Perdomo, G. Hepatic insulin clearance: Mechanism and physiology. Physiology 2019, 34, 198–215.
  8. Wu, T.; Rayner, C.K.; Jones, K.; Horowitz, M. Dietary effects on incretin hormone secretion. Vitam. Horm. 2010, 84, 81–110.
  9. Guo, C.; Huang, T.; Chen, A.; Chen, X.; Wang, L.; Shen, F.; Gu, X. Glucagon-like peptide 1 improves insulin resistance in vitro through anti-inflammation of macrophages. Braz. J. Med Biol. Res. 2016, 49, e5826.
  10. Jiang, Y.; Wang, Z.; Ma, B.; Fan, L.; Yi, N.; Lu, B.; Wang, Q.; Liu, R. GLP-1 improves adipocyte insulin sensitivity following induction of endoplasmic reticulum stress. Front. Pharmacol. 2018, 9, 1168.
  11. Gao, H.; Wang, X.; Zhang, Z.; Yang, Y.; Yang, J.; Li, X.; Ning, G. GLP-1 amplifies insulin signaling by up-regulation of IRβ, IRS-1 and Glut4 in 3T3-L1 adipocytes. Endocrine 2007, 32, 90–95.
  12. MacDonald, P.E.; El-Kholy, W.; Riedel, M.J.; Salapatek, A.M.F.; Light, P.E.; Wheeler, M.B. The multiple actions of GLP-1 on the process of glucose-stimulated insulin secretion. Diabetes 2002, 51 (Suppl. S3), S434–S442.
  13. Muscelli, E.; Mari, A.; Casolaro, A.; Camastra, S.; Seghieri, G.; Gastaldelli, A.; Holst, J.J.; Ferrannini, E. Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 2008, 57, 1340–1348.
  14. Ranganath, L.; Beety, J.; Morgan, L.; Wright, J.; Howland, R.; Marks, V. Attenuated GLP-1 secretion in obesity: Cause or consequence? Gut 1996, 38, 916–919.
  15. Lynch, C.J.; Adams, S.H. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat. Rev. Endocrinol. 2014, 10, 723–736.
  16. Gao, Z.; Hwang, D.; Bataille, F.; Lefevre, M.; York, D.; Quon, M.J.; Ye, J. Serine phosphorylation of insulin receptor substrate 1 by inhibitor κB kinase complex. J. Biol. Chem. 2002, 277, 48115–48121.
  17. Zhang, J.; Gao, Z.; Yin, J.; Quon, M.J.; Ye, J. S6K directly phosphorylates IRS-1 on Ser-270 to promote insulin resistance in response to TNF-α signaling through IKK2. J. Biol. Chem. 2008, 283, 35375–35382.
  18. Cai, D.; Yuan, M.; Frantz, D.F.; Melendez, P.A.; Hansen, L.; Lee, J.; Shoelson, S.E. Local and systemic insulin resistance resulting from hepatic activation of IKK-β and NF-κB. Nat. Med. 2005, 11, 183–190.
  19. Qi, Y.; Xu, Z.; Zhu, Q.; Thomas, C.; Kumar, R.; Feng, H.; Dostal, D.E.; White, M.F.; Baker, K.M.; Guo, S. Myocardial loss of IRS1 and IRS2 causes heart failure and is controlled by p38α MAPK during insulin resistance. Diabetes 2013, 62, 3887–3900.
  20. Lee, M.; Kim, M.; Park, J.S.; Lee, S.; You, J.; Ahn, C.W.; Kim, K.R.; Kang, S. Higher glucagon-to-insulin ratio is associated with elevated glycated hemoglobin levels in type 2 diabetes patients. Korean J. Intern. Med. 2019, 34, 1068.
  21. Saltiel, A.R.; Kahn, C.R. Insulin signalling and the regulation of glucose and lipid metabolism. Nature 2001, 414, 799–806.
  22. Boucher, J.; Kleinridders, A.; Kahn, C.R. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb. Perspect. Biol. 2014, 6, a009191.
  23. Nagao, H.; Jayavelu, A.K.; Cai, W.; Pan, H.; Dreyfuss, J.M.; Batista, T.M.; Brandão, B.B.; Mann, M.; Kahn, C.R. Unique ligand and kinase-independent roles of the insulin receptor in regulation of cell cycle, senescence and apoptosis. Nat. Commun. 2023, 14, 57.
  24. Leney, S.E.; Tavaré, J.M. The molecular basis of insulin-stimulated glucose uptake: Signalling, trafficking and potential drug targets. J. Endocrinol. 2009, 203, 1–18.
  25. Sano, H.; Eguez, L.; Teruel, M.N.; Fukuda, M.; Chuang, T.D.; Chavez, J.A.; Lienhard, G.E.; McGraw, T.E. Rab10, a target of the AS160 Rab GAP, is required for insulin-stimulated translocation of GLUT4 to the adipocyte plasma membrane. Cell Metab. 2007, 5, 293–303.
  26. Eguez, L.; Lee, A.; Chavez, J.A.; Miinea, C.P.; Kane, S.; Lienhard, G.E.; McGraw, T.E. Full intracellular retention of GLUT4 requires AS160 Rab GTPase activating protein. Cell Metab. 2005, 2, 263–272.
  27. Larance, M.; Ramm, G.; Stockli, J.; van Dam, E.M.; Winata, S.; Wasinger, V.; Simpson, F.; Graham, M.; Junutula, J.R.; Guilhaus, M. Characterization of the role of the Rab GTPase-activating protein AS160 in insulin-regulated GLUT4 trafficking. J. Biol. Chem. 2005, 280, 37803–37813.
  28. Ikonomov, O.C.; Sbrissa, D.; Mlak, K.; Shisheva, A. Requirement for PIKfyve enzymatic activity in acute and long-term insulin cellular effects. Endocrinology 2002, 143, 4742–4754.
  29. Ikonomov, O.C.; Sbrissa, D.; Dondapati, R.; Shisheva, A. ArPIKfyve–PIKfyve interaction and role in insulin-regulated GLUT4 translocation and glucose transport in 3T3-L1 adipocytes. Exp. Cell Res. 2007, 313, 2404–2416.
  30. Berwick, D.C.; Dell, G.C.; Welsh, G.I.; Heesom, K.J.; Hers, I.; Fletcher, L.M.; Cooke, F.T.; Tavare, J.M. Protein kinase B phosphorylation of PIKfyve regulates the trafficking of GLUT4 vesicles. J. Cell Sci. 2004, 117, 5985–5993.
  31. Cross, D.A.; Alessi, D.R.; Cohen, P.; Andjelkovich, M.; Hemmings, B.A. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 1995, 378, 785–789.
  32. Dong, X.C.; Copps, K.D.; Guo, S.; Li, Y.; Kollipara, R.; DePinho, R.A.; White, M.F. Inactivation of hepatic Foxo1 by insulin signaling is required for adaptive nutrient homeostasis and endocrine growth regulation. Cell Metab. 2008, 8, 65–76.
  33. Schmoll, D.; Walker, K.S.; Alessi, D.R.; Grempler, R.; Burchell, A.; Guo, S.; Walther, R.; Unterman, T.G. Regulation of glucose-6-phosphatase gene expression by protein kinase Balpha and the forkhead transcription factor FKHR. Evidence for insulin response unit-dependent and -independent effects of insulin on promoter activity. J. Biol. Chem. 2000, 275, 36324–36333.
  34. Rena, G.; Guo, S.; Cichy, S.C.; Unterman, T.G.; Cohen, P. Phosphorylation of the transcription factor forkhead family member FKHR by protein kinase B. J. Biol. Chem. 1999, 274, 17179–17183.
  35. Guo, S.; Rena, G.; Cichy, S.; He, X.; Cohen, P.; Unterman, T. Phosphorylation of serine 256 by protein kinase B disrupts transactivation by FKHR and mediates effects of insulin on insulin-like growth factor-binding protein-1 promoter activity through a conserved insulin response sequence. J. Biol. Chem. 1999, 274, 17184–17192.
  36. Yoon, N.A.; Diano, S. Hypothalamic glucose-sensing mechanisms. Diabetologia 2021, 64, 985–993.
  37. Könner, A.C.; Janoschek, R.; Plum, L.; Jordan, S.D.; Rother, E.; Ma, X.; Xu, C.; Enriori, P.; Hampel, B.; Barsh, G.S. Insulin action in AgRP-expressing neurons is required for suppression of hepatic glucose production. Cell Metab. 2007, 5, 438–449.
  38. Inoue, H.; Ogawa, W.; Asakawa, A.; Okamoto, Y.; Nishizawa, A.; Matsumoto, M.; Teshigawara, K.; Matsuki, Y.; Watanabe, E.; Hiramatsu, R. Role of hepatic STAT3 in brain-insulin action on hepatic glucose production. Cell Metab. 2006, 3, 267–275.
  39. Gelling, R.; Du, X.; Dichmann, D.; Rømer, J.; Huang, H.; Cui, L.; Obici, S.; Tang, B.; Holst, J.; Fledelius, C. Lower blood glucose, hyperglucagonemia, and pancreatic α cell hyperplasia in glucagon receptor knockout mice. Proc. Natl. Acad. Sci. USA 2003, 100, 1438–1443.
  40. Cheng, X.; Ji, Z.; Tsalkova, T.; Mei, F. Epac and PKA: A tale of two intracellular cAMP receptors. Acta Biochim. Biophys. Sin. 2008, 40, 651–662.
  41. Rui, L. Energy metabolism in the liver. Compr. Physiol. 2014, 4, 177.
  42. Oh, K.-J.; Han, H.-S.; Kim, M.-J.; Koo, S.-H. CREB and FoxO1: Two transcription factors for the regulation of hepatic gluconeogenesis. BMB Rep. 2013, 46, 567.
  43. Koo, S.-H.; Flechner, L.; Qi, L.; Zhang, X.; Screaton, R.A.; Jeffries, S.; Hedrick, S.; Xu, W.; Boussouar, F.; Brindle, P. The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 2005, 437, 1109.
  44. Xu, W.; Kasper, L.H.; Lerach, S.; Jeevan, T.; Brindle, P.K. Individual CREB-target genes dictate usage of distinct cAMP-responsive coactivation mechanisms. EMBO J. 2007, 26, 2890–2903.
  45. Wu, Y.; Pan, Q.; Yan, H.; Zhang, K.; Guo, X.; Xu, Z.; Yang, W.; Qi, Y.; Guo, C.A.; Hornsby, C. Novel mechanism of Foxo1 phosphorylation in glucagon signaling in control of glucose homeostasis. Diabetes 2018, 67, 2167–2182.
  46. Yang, W.; Liao, W.; Li, X.; Ai, W.; Pan, Q.; Shen, Z.; Jiang, W.; Guo, S. Hepatic p38α MAPK controls gluconeogenesis via FOXO1 phosphorylation at S273 during glucagon signalling in mice. Diabetologia 2023, 66, 1322–1339.
  47. Zhang, K.; Li, L.; Qi, Y.; Zhu, X.; Gan, B.; DePinho, R.A.; Averitt, T.; Guo, S. Hepatic suppression of Foxo1 and Foxo3 causes hypoglycemia and hyperlipidemia in mice. Endocrinology 2012, 153, 631–646.
  48. Haeusler, R.A.; Kaestner, K.H.; Accili, D. FoxOs function synergistically to promote glucose production. J. Biol. Chem. 2010, 285, 35245–35248.
  49. Matsuzaki, H.; Daitoku, H.; Hatta, M.; Tanaka, K.; Fukamizu, A. Insulin-induced phosphorylation of FKHR (Foxo1) targets to proteasomal degradation. Proc. Natl. Acad. Sci. USA 2003, 100, 11285–11290.
  50. Zhang, K.; Guo, X.; Yan, H.; Wu, Y.; Pan, Q.; Shen, J.Z.; Li, X.; Chen, Y.; Li, L.; Qi, Y. Phosphorylation of forkhead protein FoxO1 at S253 regulates glucose homeostasis in mice. Endocrinology 2019, 160, 1333–1347.
  51. Yang, W.; Kim, D.M.; Jiang, W.; Ai, W.; Pan, Q.; Rahman, S.; Cai, J.J.; Brashear, W.A.; Sun, Y.; Guo, S. Suppression of FOXO1 attenuates inflamm-aging and improves liver function during aging. Aging Cell 2023, 22, e13968.
  52. Cheng, Z.; Guo, S.; Copps, K.; Dong, X.; Kollipara, R.; Rodgers, J.T.; Depinho, R.A.; Puigserver, P.; White, M.F. Foxo1 integrates insulin signaling with mitochondrial function in the liver. Nat. Med. 2009, 15, 1307–1311.
  53. Yang, W.; Yan, H.; Pan, Q.; Shen, J.Z.; Zhou, F.; Wu, C.; Sun, Y.; Guo, S. Glucagon regulates hepatic mitochondrial function and biogenesis through FOXO1. J. Endocrinol. 2019, 241, 265–278.
  54. Liao, W.; Yang, W.; Shen, Z.; Ai, W.; Pan, Q.; Sun, Y.; Guo, S. Heme Oxygenase-1 Regulates Ferrous Iron and Foxo1 in Control of Hepatic Gluconeogenesis. Diabetes 2021, 70, 696–709.
  55. Pan, Q.; Ai, W.; Chen, Y.; Kim, D.M.; Shen, Z.; Yang, W.; Jiang, W.; Sun, Y.; Safe, S.; Guo, S. Reciprocal regulation of hepatic TGF-β1 and Foxo1 controls gluconeogenesis and energy expenditure. Diabetes 2023, 72, 1193–1206.
  56. Pan, Q.; Gao, M.; Kim, D.; Ai, W.; Yang, W.; Jiang, W.; Brashear, W.; Dai, Y.; Li, S.; Sun, Y. Hepatocyte FoxO1 deficiency protects from liver fibrosis via reducing inflammation and TGF-β1 mediated HSC activation. Cell. Mol. Gastroenterol. Hepatol. 2023.
  57. Laville, M.; Nazare, J.A. Diabetes, insulin resistance and sugars. Obes. Rev. 2009, 10, 24–33.
  58. Stanhope, K.L.; Schwarz, J.M.; Keim, N.L.; Griffen, S.C.; Bremer, A.A.; Graham, J.L.; Hatcher, B.; Cox, C.L.; Dyachenko, A.; Zhang, W. Consuming fructose-sweetened, not glucose-sweetened, beverages increases visceral adiposity and lipids and decreases insulin sensitivity in overweight/obese humans. J. Clin. Investig. 2009, 119, 1322–1334.
  59. Lecoultre, V.; Egli, L.; Carrel, G.; Theytaz, F.; Kreis, R.; Schneiter, P.; Boss, A.; Zwygart, K.; Lê, K.A.; Bortolotti, M. Effects of fructose and glucose overfeeding on hepatic insulin sensitivity and intrahepatic lipids in healthy humans. Obesity 2013, 21, 782–785.
  60. Heden, T.D.; Liu, Y.; Park, Y.-M.; Nyhoff, L.M.; Winn, N.C.; Kanaley, J.A. Moderate amounts of fructose-or glucose-sweetened beverages do not differentially alter metabolic health in male and female adolescents. Am. J. Clin. Nutr. 2014, 100, 796–805.
  61. Cha, S.H.; Wolfgang, M.; Tokutake, Y.; Chohnan, S.; Lane, M.D. Differential effects of central fructose and glucose on hypothalamic malonyl–CoA and food intake. Proc. Natl. Acad. Sci. USA 2008, 105, 16871–16875.
  62. Teff, K.L.; Elliott, S.S.; Tschöp, M.; Kieffer, T.J.; Rader, D.; Heiman, M.; Townsend, R.R.; Keim, N.L.; D’alessio, D.; Havel, P.J. Dietary fructose reduces circulating insulin and leptin, attenuates postprandial suppression of ghrelin, and increases triglycerides in women. J. Clin. Endocrinol. Metab. 2004, 89, 2963–2972.
  63. Miller, C.C.; Martin, R.J.; Whitney, M.L.; Edwards, G.L. Intracerebroventricular injection of fructose stimulates feeding in rats. Nutr. Neurosci. 2002, 5, 359–362.
  64. Stanhope, K.L. Sugar consumption, metabolic disease and obesity: The state of the controversy. Crit. Rev. Clin. Lab. Sci. 2016, 53, 52–67.
  65. Tappy, L.; Lê, K.-A. Metabolic effects of fructose and the worldwide increase in obesity. Physiol. Rev. 2010, 90, 23–46.
  66. Zhang, C.; Chen, X.; Zhu, R.-M.; Zhang, Y.; Yu, T.; Wang, H.; Zhao, H.; Zhao, M.; Ji, Y.-L.; Chen, Y.-H. Endoplasmic reticulum stress is involved in hepatic SREBP-1c activation and lipid accumulation in fructose-fed mice. Toxicol. Lett. 2012, 212, 229–240.
  67. Kim, M.-S.; Krawczyk, S.A.; Doridot, L.; Fowler, A.J.; Wang, J.X.; Trauger, S.A.; Noh, H.-L.; Kang, H.J.; Meissen, J.K.; Blatnik, M. ChREBP regulates fructose-induced glucose production independently of insulin signaling. J. Clin. Investig. 2019, 126, 4372–4386.
  68. Rao, S.S.; Attaluri, A.; Anderson, L.; Stumbo, P. Ability of the normal human small intestine to absorb fructose: Evaluation by breath testing. Clin. Gastroenterol. Hepatol. 2007, 5, 959–963.
  69. Wang, Y.; Qi, W.; Song, G.; Pang, S.; Peng, Z.; Li, Y.; Wang, P. High-fructose diet increases inflammatory cytokines and alters gut microbiota composition in rats. Mediat. Inflamm. 2020, 2020, 6672636.
  70. Do, M.H.; Lee, E.; Oh, M.-J.; Kim, Y.; Park, H.-Y. High-glucose or-fructose diet cause changes of the gut microbiota and metabolic disorders in mice without body weight change. Nutrients 2018, 10, 761.
  71. Todoric, J.; Di Caro, G.; Reibe, S.; Henstridge, D.C.; Green, C.R.; Vrbanac, A.; Ceteci, F.; Conche, C.; McNulty, R.; Shalapour, S. Fructose stimulated de novo lipogenesis is promoted by inflammation. Nat. Metab. 2020, 2, 1034–1045.
  72. Uysal, K.T.; Wiesbrock, S.M.; Marino, M.W.; Hotamisligil, G.S. Protection from obesity-induced insulin resistance in mice lacking TNF-α function. Nature 1997, 389, 610–614.
  73. Johnston, C.A.; Stevens, B.; Foreyt, J.P. The role of low-calorie sweeteners in diabetes. Eur. Endocrinol. 2013, 9, 96.
  74. Miller, P.E.; Perez, V. Low-calorie sweeteners and body weight and composition: A meta-analysis of randomized controlled trials and prospective cohort studies. Am. J. Clin. Nutr. 2014, 100, 765–777.
  75. Azad, M.B.; Abou-Setta, A.M.; Chauhan, B.F.; Rabbani, R.; Lys, J.; Copstein, L.; Mann, A.; Jeyaraman, M.M.; Reid, A.E.; Fiander, M. Nonnutritive sweeteners and cardiometabolic health: A systematic review and meta-analysis of randomized controlled trials and prospective cohort studies. Cmaj 2017, 189, E929–E939.
  76. Schulze, M.B.; Schulz, M.; Heidemann, C.; Schienkiewitz, A.; Hoffmann, K.; Boeing, H. Fiber and magnesium intake and incidence of type 2 diabetes: A prospective study and meta-analysis. Arch. Intern. Med. 2007, 167, 956–965.
  77. Robertson, M.D.; Bickerton, A.S.; Dennis, A.L.; Vidal, H.; Frayn, K.N. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am. J. Clin. Nutr. 2005, 82, 559–567.
  78. Weickert, M.O.; Möhlig, M.; Schöfl, C.; Arafat, A.M.; Otto, B.; Viehoff, H.; Koebnick, C.; Kohl, A.; Spranger, J.; Pfeiffer, A.F. Cereal fiber improves whole-body insulin sensitivity in overweight and obese women. Diabetes Care 2006, 29, 775–780.
  79. Pereira, M.A.; Jacobs, D.R., Jr.; Pins, J.J.; Raatz, S.K.; Gross, M.D.; Slavin, J.L.; Seaquist, E.R. Effect of whole grains on insulin sensitivity in overweight hyperinsulinemic adults. Am. J. Clin. Nutr. 2002, 75, 848–855.
  80. Hanai, H.; Ikuma, M.; Sato, Y.; Iida, T.; Hosoda, Y.; Matsushita, I.; Nogaki, A.; Yamada, M.; Kaneko, E. Long-term effects of water-soluble corn bran hemicellulose on glucose tolerance in obese and non-obese patients: Improved insulin sensitivity and glucose metabolism in obese subjects. Biosci. Biotechnol. Biochem. 1997, 61, 1358–1361.
  81. Sierra, M.; Garcia, J.; Fernández, N.; Diez, M.; Calle, A.; Sahagun, A. Effects of ispaghula husk and guar gum on postprandial glucose and insulin concentrations in healthy subjects. Eur. J. Clin. Nutr. 2001, 55, 235–243.
  82. Isken, F.; Klaus, S.; Osterhoff, M.; Pfeiffer, A.F.; Weickert, M.O. Effects of long-term soluble vs. insoluble dietary fiber intake on high-fat diet-induced obesity in C57BL/6J mice. J. Nutr. Biochem. 2010, 21, 278–284.
  83. Brooks, L.; Viardot, A.; Tsakmaki, A.; Stolarczyk, E.; Howard, J.K.; Cani, P.D.; Everard, A.; Sleeth, M.L.; Psichas, A.; Anastasovskaj, J. Fermentable carbohydrate stimulates FFAR2-dependent colonic PYY cell expansion to increase satiety. Mol. Metab. 2017, 6, 48–60.
  84. Cani, P.D.; Knauf, C.; Iglesias, M.A.; Drucker, D.J.; Delzenne, N.M.; Burcelin, R. Improvement of glucose tolerance and hepatic insulin sensitivity by oligofructose requires a functional glucagon-like peptide 1 receptor. Diabetes 2006, 55, 1484–1490.
  85. Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein–coupled receptor FFAR2. Diabetes 2012, 61, 364–371.
  86. Guo, X.; Li, X.; Yang, W.; Liao, W.; Shen, J.Z.; Ai, W.; Pan, Q.; Sun, Y.; Zhang, K.; Zhang, R.; et al. Metformin Targets Foxo1 to Control Glucose Homeostasis. Biomolecules 2021, 11, 873.
  87. Gulliford, M.; Ukoumunne, O. Determinants of glycated haemoglobin in the general population: Associations with diet, alcohol and cigarette smoking. Eur. J. Clin. Nutr. 2001, 55, 615–623.
  88. Mayer, E.J.; Newman, B.; Quesenberry, C.P., Jr.; Selby, J.V. Usual dietary fat intake and insulin concentrations in healthy women twins. Diabetes Care 1993, 16, 1459–1469.
  89. Harding, A.-H.; Sargeant, L.A.; Welch, A.; Oakes, S.; Luben, R.N.; Bingham, S.; Day, N.E.; Khaw, K.-T.; Wareham, N.J. Fat consumption and HbA1c levels: The EPIC-Norfolk study. Diabetes Care 2001, 24, 1911–1916.
  90. Marshall, J.A.; Hoag, S.; Shetterly, S.; Hamman, R.F. Dietary fat predicts conversion from impaired glucose tolerance to NIDDM: The San Luis Valley Diabetes Study. Diabetes Care 1994, 17, 50–56.
  91. Glass, C.K.; Olefsky, J.M. Inflammation and lipid signaling in the etiology of insulin resistance. Cell Metab. 2012, 15, 635–645.
  92. Vessby, B.; Uusitupa, M.; Hermansen, K.; Riccardi, G.; Rivellese, A.A.; Tapsell, L.C.; Nälsén, C.; Berglund, L.; Louheranta, A.; Rasmussen, B. Substituting dietary saturated for monounsaturated fat impairs insulin sensitivity in healthy men and women: The KANWU Study. Diabetologia 2001, 44, 312–319.
  93. Clarke, S.D. Polyunsaturated fatty acid regulation of gene transcription: A mechanism to improve energy balance and insulin resistance. Br. J. Nutr. 2000, 83, S59–S66.
  94. Ramel, A.; Martinez, A.; Kiely, M.; Morais, G.; Bandarra, N.; Thorsdottir, I. Beneficial effects of long-chain n-3 fatty acids included in an energy-restricted diet on insulin resistance in overweight and obese European young adults. Diabetologia 2008, 51, 1261–1268.
  95. Huang, T.; Wahlqvist, M.L.; Xu, T.; Xu, A.; Zhang, A.; Li, D. Increased plasma n-3 polyunsaturated fatty acid is associated with improved insulin sensitivity in type 2 diabetes in China. Mol. Nutr. Food Res. 2010, 54, S112–S119.
  96. Lee, J.Y.; Plakidas, A.; Lee, W.H.; Heikkinen, A.; Chanmugam, P.; Bray, G.; Hwang, D.H. Differential modulation of Toll-like receptors by fatty acids: Preferential inhibition by n-3 polyunsaturated fatty acids. J. Lipid Res. 2003, 44, 479–486.
  97. Talukdar, S.; Bae, E.J.; Imamura, T.; Morinaga, H.; Fan, W.; Li, P.; Lu, W.J.; Watkins, S.M.; Olefsky, J.M. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 2010, 142, 687–698.
  98. Feskens, E.J.; Bowles, C.H.; Kromhout, D. Inverse association between fish intake and risk of glucose intolerance in normoglycemic elderly men and women. Diabetes Care 1991, 14, 935–941.
  99. Halton, T.L.; Hu, F.B. The effects of high protein diets on thermogenesis, satiety and weight loss: A critical review. J. Am. Coll. Nutr. 2004, 23, 373–385.
  100. Hu, F.B. Protein, body weight, and cardiovascular health. Am. J. Clin. Nutr. 2005, 82, 242S–247S.
  101. Mithieux, G.; Misery, P.; Magnan, C.; Pillot, B.; Gautier-Stein, A.; Bernard, C.; Rajas, F.; Zitoun, C. Portal sensing of intestinal gluconeogenesis is a mechanistic link in the diminution of food intake induced by diet protein. Cell Metab. 2005, 2, 321–329.
  102. Penhoat, A.; Mutel, E.; Amigo-Correig, M.; Pillot, B.; Stefanutti, A.; Rajas, F.; Mithieux, G. Protein-induced satiety is abolished in the absence of intestinal gluconeogenesis. Physiol. Behav. 2011, 105, 89–93.
  103. Spiller, G.A.; Jensen, C.D.; Pattison, T.; Chuck, C.S.; Whittam, J.H.; Scala, J. Effect of protein dose on serum glucose and insulin response to sugars. Am. J. Clin. Nutr. 1987, 46, 474–480.
  104. Wu, T.; Little, T.J.; Bound, M.J.; Borg, M.; Zhang, X.; Deacon, C.F.; Horowitz, M.; Jones, K.L.; Rayner, C.K. A protein preload enhances the glucose-lowering efficacy of vildagliptin in type 2 diabetes. Diabetes Care 2016, 39, 511–517.
  105. Ma, J.; Stevens, J.E.; Cukier, K.; Maddox, A.F.; Wishart, J.M.; Jones, K.L.; Clifton, P.M.; Horowitz, M.; Rayner, C.K. Effects of a protein preload on gastric emptying, glycemia, and gut hormones after a carbohydrate meal in diet-controlled type 2 diabetes. Diabetes Care 2009, 32, 1600–1602.
  106. Gannon, M.C.; Nuttall, F.Q.; Saeed, A.; Jordan, K.; Hoover, H. An increase in dietary protein improves the blood glucose response in persons with type 2 diabetes. Am. J. Clin. Nutr. 2003, 78, 734–741.
  107. Linn, T.; Santosa, B.; Grönemeyer, D.; Aygen, S.; Scholz, N.; Busch, M.; Bretzel, R. Effect of long-term dietary protein intake on glucose metabolism in humans. Diabetologia 2000, 43, 1257–1265.
  108. Linn, T.; Geyer, R.; Prassek, S.; Laube, H. Effect of dietary protein intake on insulin secretion and glucose metabolism in insulin-dependent diabetes mellitus. J. Clin. Endocrinol. Metab. 1996, 81, 3938–3943.
  109. Newgard, C.B.; An, J.; Bain, J.R.; Muehlbauer, M.J.; Stevens, R.D.; Lien, L.F.; Haqq, A.M.; Shah, S.H.; Arlotto, M.; Slentz, C.A. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009, 9, 311–326.
  110. Krebs, M.; Krssak, M.; Bernroider, E.; Anderwald, C.; Brehm, A.; Meyerspeer, M.; Nowotny, P.; Roth, E.; Waldhäusl, W.; Roden, M. Mechanism of amino acid-induced skeletal muscle insulin resistance in humans. Diabetes 2002, 51, 599–605.
  111. Tremblay, F.; Krebs, M.; Dombrowski, L.; Brehm, A.; Bernroider, E.; Roth, E.; Nowotny, P.; Waldhäusl, 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, 2674–2684.
  112. Tremblay, F.; Marette, A. Amino acid and insulin signaling via the mTOR/p70 S6 kinase pathway: A negative feedback mechanism leading to insulin resistance in skeletal muscle cells. J. Biol. Chem. 2001, 276, 38052–38060.
  113. Takano, A.; Usui, I.; Haruta, T.; Kawahara, J.; Uno, T.; Iwata, M.; Kobayashi, M. Mammalian target of rapamycin pathway regulates insulin signaling via subcellular redistribution of insulin receptor substrate 1 and integrates nutritional signals and metabolic signals of insulin. Mol. Cell. Biol. 2001, 21, 5050–5062.
  114. Patti, M.-E.; Brambilla, E.; Luzi, L.; Landaker, E.J.; Kahn, C.R. Bidirectional modulation of insulin action by amino acids. J. Clin. Investig. 1998, 101, 1519–1529.
  115. Zhao, H.; Zhang, F.; Sun, D.; Wang, X.; Zhang, X.; Zhang, J.; Yan, F.; Huang, C.; Xie, H.; Lin, C. Branched-chain amino acids exacerbate obesity-related hepatic glucose and lipid metabolic disorders via attenuating Akt2 signaling. Diabetes 2020, 69, 1164–1177.
  116. Würtz, P.; Soininen, P.; Kangas, A.J.; Rönnemaa, T.; Lehtimäki, T.; Kähönen, M.; Viikari, J.S.; Raitakari, O.T.; Ala-Korpela, M. Branched-chain and aromatic amino acids are predictors of insulin resistance in young adults. Diabetes Care 2013, 36, 648–655.
  117. Phielix, E.; Jelenik, T.; Nowotny, P.; Szendroedi, J.; Roden, M. Reduction of non-esterified fatty acids improves insulin sensitivity and lowers oxidative stress, but fails to restore oxidative capacity in type 2 diabetes: A randomised clinical trial. Diabetologia 2014, 57, 572–581.
  118. Vanweert, F.; Schrauwen, P.; Phielix, E. Role of branched-chain amino acid metabolism in the pathogenesis of obesity and type 2 diabetes-related metabolic disturbances BCAA metabolism in type 2 diabetes. Nutr. Diabetes 2022, 12, 35.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 151
Revisions: 2 times (View History)
Update Date: 19 Dec 2023
1000/1000
Video Production Service