The Role of Palmitoleic Acid in Regulating Hepatic Gluconeogenesis through SIRT3 in Obese Mice

Xin Guo, Xiaofan Jiang, Keyun Chen, Qijian Liang, Shixiu Zhang, Juan Zheng, Xiaomin Ma, Hongmei Jiang, Hao Wu, Qiang Tong, Xin Guo, Xiaofan Jiang, Keyun Chen, Qijian Liang, Shixiu Zhang, Juan Zheng, Xiaomin Ma, Hongmei Jiang, Hao Wu, Qiang Tong

Abstract

Hepatic gluconeogenesis is a crucial process to maintain glucose level during starvation. However, unabated glucose production in diabetic patients is a major contributor to hyperglycemia. Palmitoleic acid is a monounsaturated fatty acid (16:1n7) that is available from dietary sources. Palmitoleic acid exhibits health beneficial effects on diabetes, insulin resistance, inflammation, and metabolic syndrome. However, the mechanism by which palmitoleate reduces blood glucose is still unclear. SIRT3 is a key metabolism-regulating NAD+-dependent protein deacetylase. It is known that fasting elevates the expression of SIRT3 in the liver and it regulates many aspects of liver's response to nutrient deprivation, such as fatty acid oxidation and ketone body formation. However, it is unknown whether SIRT3 also regulates gluconeogenesis. Our study revealed that palmitoleic acid reduced hepatic gluconeogenesis and the expression of SIRT3 under high-fat diet conditions. Overexpression of SIRT3 in the liver and hepatocytes enhanced gluconeogenesis. Further study revealed that SIRT3 played a role in enhancing the activities of gluconeogenic enzymes, such as PEPCK, PC, and MDH2. Therefore, our study indicated that under a high-fat diet, palmitoleic acid decreased gluconeogenesis by reducing enzymatic activities of PEPCK, PC, and MDH2 by down-regulating the expression of SIRT3.

Keywords: SIRT3; gluconeogenesis; high-fat diet; palmitoleic acid.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Palmitoleic acid increases systemic glucose clearance and reduces hepatic gluconeogenesis in obese mice. C57BL/6N male mice of 5–6 weeks of age were fed with LFD (low-fat diet) or HFD (high-fat diet) for 12 weeks. At the 13th week, BSA (bovine serum albumin), oleic acid, or palmitoleic acid was administered intragastrically. At the15th, 16th, and 17th weeks, GTT (Glucose tolerance tests), ITT (insulin tolerance tests) and PTT (pyruvate tolerance) were performed, respectively. (A) GTT for LFD group. (B) Area under curve of GTT in LFD group. (C) GTT for HFD group. (D) Area under curve of GTT in HFD group. (E) ITT for LFD group. (F) Area under curve of ITT in LFD group. (G) ITT for HFD group. (H) Area under curve of ITT in HFD group. (I) PTT for LFD group. (J) Area under curve of PTT in LFD group. (K) PTT for HFD group. (L) Area under curve of PTT in HFD group. Above data: n = 6–13 mice per group. (M) HOMA-IR in LFD group. (N) HOMA-IR in HFD group. Calculated based on fasting glucose level (mmol/L) and fasting insulin level (microU/mL). For HOMA-IR: n = 4–8 mice per group. The data are mean ± s.e. (error bars). *, p < 0.05, **, p < 0.01, ***, p < 0.001, BSA vs. palmitoleic acid; †, p< 0.05, ††, p < 0.01, †††, p < 0.001, oleic acid vs. palmitoleic acid; ‡, p < 0.05, ‡‡, p < 0.01, BSA vs. oleic acid.
Figure 1
Figure 1
Palmitoleic acid increases systemic glucose clearance and reduces hepatic gluconeogenesis in obese mice. C57BL/6N male mice of 5–6 weeks of age were fed with LFD (low-fat diet) or HFD (high-fat diet) for 12 weeks. At the 13th week, BSA (bovine serum albumin), oleic acid, or palmitoleic acid was administered intragastrically. At the15th, 16th, and 17th weeks, GTT (Glucose tolerance tests), ITT (insulin tolerance tests) and PTT (pyruvate tolerance) were performed, respectively. (A) GTT for LFD group. (B) Area under curve of GTT in LFD group. (C) GTT for HFD group. (D) Area under curve of GTT in HFD group. (E) ITT for LFD group. (F) Area under curve of ITT in LFD group. (G) ITT for HFD group. (H) Area under curve of ITT in HFD group. (I) PTT for LFD group. (J) Area under curve of PTT in LFD group. (K) PTT for HFD group. (L) Area under curve of PTT in HFD group. Above data: n = 6–13 mice per group. (M) HOMA-IR in LFD group. (N) HOMA-IR in HFD group. Calculated based on fasting glucose level (mmol/L) and fasting insulin level (microU/mL). For HOMA-IR: n = 4–8 mice per group. The data are mean ± s.e. (error bars). *, p < 0.05, **, p < 0.01, ***, p < 0.001, BSA vs. palmitoleic acid; †, p< 0.05, ††, p < 0.01, †††, p < 0.001, oleic acid vs. palmitoleic acid; ‡, p < 0.05, ‡‡, p < 0.01, BSA vs. oleic acid.
Figure 2
Figure 2
Palmitoleic acid did not alter SIRT3 expression in the liver of mice under LFD. C57BL/6N male mice of 5–6 weeks of age were fed with LFD for 12 weeks. At the 13th week, BSA, oleic acid, or palmitoleic acid was administered intragastrically. At the 18th week, mice were euthanized, and liver tissue was collected. (A) Expression of SIRT3 was detected using Western blot analysis. (B) Quantification of SIRT3 expression. AU, arbitrary units. The data are mean ± s.e. (error bars). n = 5 mice per group.
Figure 3
Figure 3
Palmitoleic acid reduced the expression of SIRT3 and MDH2 but did not change the protein levels of PC, PEPCK, and SIRT1 in the liver of mice under HFD. C57BL/6N male mice of 5–6 weeks of age were fed with HFD for 12 weeks. At the 13th week, BSA, oleic acid, or palmitoleic acid were administered intragastrically. At the 18th week, mice were euthanized, and liver tissues were collected. (A) Expression of SIRT3, SIRT1, and gluconeogenic enzymes in the liver of mice under HFD. Protein expression was detected using Western blot analysis. (B) Quantification of SIRT3 protein level. (C) Quantification of PC protein level. (D) Quantification of PEPCK protein level. (E) Quantification of MDH2 protein level. (F) Quantification of SIRT1 protein level. AU, arbitrary units. The data are mean ± s.e. (error bars). n = 4–6 mice per group. *, p < 0.05, **, p < 0.01, HFD BSA vs. HFD palmitoleic acid; †, p < 0.05, oleic acid vs. palmitoleic acid.
Figure 4
Figure 4
Palmitoleic acid reduces enzymatic activities of MDH2, PC, and PEPCK under HFD. C57BL/6N male mice of 5–6 weeks of age were fed with LFD or HFD for 12 weeks. At the 13th week, BSA, oleic acid, or palmitoleic acid was administered intragastrically. At the 18th week, mice were sacrificed, and liver tissue was collected. The enzyme activities of MDH2, PC, and PEPCK in the liver were measured. (A) Enzymatic activity of MDH2 in the liver of HFD-fed mice. (B) Enzymatic activity of PC in the liver of HFD-fed mice. (C) Enzymatic activity of PEPCK in the liver of HFD-fed mice. The data are mean ± s.e. (error bars). n = 4–7 mice per group. *, p < 0.05, **, p < 0.01, HFD BSA vs. HFD palmitoleic acid; ††, p < 0.01, †††, p < 0.001, HFD oleic acid vs. HFD palmitoleic acid.
Figure 5
Figure 5
Liver-specific SIRT3 overexpression increases gluconeogenesis. C57BL/6N male mice of 5–6 weeks of age were injected with AAV-GFP or AAV-SIRT3 via a tail vein and fed with LFD or HFD for 12 weeks, respectively. At the 13th week, PTT was performed. At the 14th week of feeding, mice were euthanized, and liver tissues were collected. (A) Protein level of SIRT3 in liver. (B) RNA level of SIRT3 in liver. (C) Protein level of SIRT3 in adipose tissue. (D) Quantification of SIRT3 protein level in adipose tissue. (E) Protein level of SIRT3 in muscle. (F) Quantification of SIRT3 protein level in muscle. The data are mean ± s.e. (error bars). n = 6 mice per group. **, p < 0.01, AAV-GFP vs. AAV-SIRT3. (G) PTT. Mice were fasted overnight and intraperitoneally injected with pyruvate (2 g/kg). Blood glucose levels were measured before injection and 15, 30, 60, and 120 min after injection. (H) Area under curve of PTT. The data are mean ± s.e. (error bars). n = 6–9 mice per group. §, p < 0.05, LFD AAV-SIRT3 vs. HFD AAV-SIRT3; *, p < 0.05, **, p < 0.01, ***, p < 0.001, LFD AAV-GFP vs. HFD AAV-GFP; ‡, p < 0.05, ‡‡‡, p < 0.001, LFD AAV-GFP vs. LFD AAV-SIRT3; †, p < 0.05, ††, p < 0.01, HFD AAV-GFP vs. HFD AAV-SIRT3.
Figure 6
Figure 6
Liver-specific SIRT3 overexpression increases MDH2 protein levels in mice. C57BL/6N male mice of 5–6 weeks of age were injected with AAV-GFP or AAV-SIRT3 via a tail vein and fed with LFD or HFD for 12 weeks. At the 14th week of feeding, mice were euthanized, and the liver tissues were collected. (A) Expression of SIRT3 and the levels of gluconeogenic-related enzymes in the liver. (B) Quantitative the expression of PEPCK protein level. (C) Quantitative the expression of PC protein level. (D) Quantification the expression of MDH2 protein level. AU, arbitrary units. The data are mean ± s.e. (error bars). n = 6 mice per group. *, p < 0.05, LFD AAV-GFP vs. HFD AAV-GFP; ‡, p < 0.05, LFD AAV-GFP vs. LFD AAV-SIRT3.
Figure 7
Figure 7
SIRT3 overexpression increases the activities of gluconeogenic enzymes. C57BL/6N male mice of 5–6 weeks of age were injected with AAV-GFP or AAV-SIRT3 via a tail vein and fed with LFD or HFD for 12 weeks. At the 14th week of feeding, mice were euthanized, and the liver tissues were collected. (A) Enzymatic activity of MDH2. (B) Enzymatic activity of PC. (C) Enzymatic activity of PEPCK. The data are mean ± s.e. (error bars). n = 4–7 mice per group *, p < 0.05, LFD AAV-GFP vs. HFD AAV-GFP; ‡, p < 0.05, ‡‡, p < 0.01, LFD AAV-GFP vs. LFD AAV-SIRT3; †, p < 0.05, ††, p < 0.01, HFD AAV-GFP vs. HFD AAV-SIRT3.
Figure 8
Figure 8
SIRT3 overexpression increases glucose production and the protein level of MDH2 in primary hepatocytes. Primary hepatocytes from HFD or LFD mice were isolated and infected with Ad-GFP or Ad-SIRT3 adenovirus. (A) Infection status after 48 h as fluorescent detection of GFP. (B) Glucose production from primary hepatocytes. In the presence of substrates (20 mM lactic acid and 2 mM pyruvate), the primary hepatocytes were stimulated with 100 nM glucagon for 3 h with or without 50 uM 3-TYP treatment for 24 h. a, p < 0.05, LFD Ad-GFP vs. LFD Ad-GFP+3-TYP; b, p < 0.05, LFD Ad-GFP vs. LFD Ad-SIRT3; c, p < 0.05, LFD Ad-GFP+3-TYP vs. LFD Ad-SIRT3; d, p <0.05, LFD Ad-GFP vs. HFD Ad-GFP; e, p < 0.05, HFD Ad-GFP vs. HFD Ad-GFP+3-TYP; f, p < 0.05, HFD Ad-GFP vs. HFD Ad-SIRT3; g, p < 0.01, HFD Ad-GFP+3-TYP vs. HFD Ad-SIRT3. (C) SIRT3, PC and MDH2 protein levels. Primary hepatocytes infected with adenovirus were treated with DMSO or 3-TYP for 24 h and 100 nM glucagon for 1 h. Cells were collected for protein extraction. Western blot analysis was used to detect the expression of SIRT3, PC, and MDH2. *, p < 0.05, LFD Ad-GFP vs. LFD Ad-SIRT3; ††, p < 0.01 HFD Ad-GFP vs. HFD Ad-SIRT3. The data are mean ± s.e. (error bars). n = 5 from three independent experiments.
Figure 9
Figure 9
Palmitoleic acid reverses the increase in gluconeogenesis caused by SIRT3 overexpression in primary hepatocytes. Primary hepatocytes from mice fed with HFD were isolated and infected with Ad-GFP or Ad-SIRT3 adenovirus. Ad-SIRT3-infected hepatocytes were also treated with palmitoleic acid for 48 h. (A) Glucose production from primary hepatocytes. In the presence of substrates (20 mM lactic acid and 2 mM pyruvate), the primary hepatocytes were stimulated with 100 nM glucagon for 3 h to detect the content of glucose in the medium. (B) SIRT3, PC, and MDH2 protein levels were detected using Western blot analysis. The data are mean ± s.e. (error bars). n = 6 from three independent experiments. *, p < 0.05, HFD Ad-GFP vs. HFD Ad-SIRT3; †, p < 0.05 HFD Ad-SIRT3 vs. HFD Ad-SIRT3+PO (palmitoleic acid).

References

    1. Naghavi M., Wang H., Lozano R., Davis A., Liang X., Zhou M., Vollset S.E., Ozgoren A.A., Abdalla S., Abd-Allah F., et al. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990-2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;385:117–171. doi: 10.1016/s0140-6736(14)61682-2.
    1. Jia G., DeMarco V.G., Sowers J.R. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Nat. Rev. Endocrinol. 2016;12:144–153. doi: 10.1038/nrendo.2015.216.
    1. Roden M., Shulman G.I. The integrative biology of type 2 diabetes. Nature. 2019;576:51–60. doi: 10.1038/s41586-019-1797-8.
    1. Hu W., Fitzgerald M., Topp B., Alam M., O’Hare T.J. A review of biological functions, health benefits, and possible de novo biosynthetic pathway of palmitoleic acid in macadamia nuts. J. Funct. Foods. 2019;62:103520. doi: 10.1016/j.jff.2019.103520.
    1. Astudillo A.M., Meana C., Guijas C., Pereira L., Lebrero P., Balboa M.A., Balsinde J. Occurrence and biological activity of palmitoleic acid isomers in phagocytic cells. J. Lipid Res. 2018;59:237–249. doi: 10.1194/jlr.M079145.
    1. Bueno-Hernandez N., Sixtos-Alonso M.S., del Pilar MilKe M., Yamamoto-Furusho J.K. Effect of Cis-palmitoleic acid supplementation on inflammation and expression of HNF4 gamma, HNF4 alpha and IL6 in patients with ulcerative colitis. Minerva Gastroenterol. E Dietol. 2017;63:257–263. doi: 10.23736/s1121-421x.17.02367-4.
    1. Talbot N.A., Wheeler-Jones C.P., Cleasby M.E. Palmitoleic acid prevents palmitic acid-induced macrophage activation and consequent p38 MAPK-mediated-skeletal muscle insulin resistance. Mol. Cell. Endocrinol. 2014;393:129–142. doi: 10.1016/j.mce.2014.06.010.
    1. Guo X., Li H., Xu H., Halim V., Zhang W., Wang H., Ong K.T., Woo S.-L., Walzem R.L., Mashek D.G., et al. Palmitoleate Induces Hepatic Steatosis but Suppresses Liver Inflammatory Response in Mice. PLoS ONE. 2012;7:e39286. doi: 10.1371/journal.pone.0039286.
    1. Bolsoni-Lopes A., Festuccia W.T., Chimin P., Farias T.S.M., Torres-Leal F.L., Cruz M.M., Andrade P.B., Hirabara S.M., Lima F.B., Alonso-Vale M.I.C. Palmitoleic acid (n-7) increases white adipocytes GLUT4 content and glucose uptake in association with AMPK activation. Lipids Health Dis. 2014;13:199. doi: 10.1186/1476-511X-13-199.
    1. Verdin E. NAD(+) in aging, metabolism, and neurodegeneration. Science. 2015;350:1208–1213. doi: 10.1126/science.aac4854.
    1. Shi T., Wang F., Stieren E., Tong Q. SIRT3, a mitochondrial sirtuin deacetylase, regulates mitochondrial function and thermogenesis in brown adipocytes. J. Biol. Chem. 2005;280:13560–13567. doi: 10.1074/jbc.M414670200.
    1. Lin L., Chen K., Khalek W.A., Ward J.L., III, Yang H., Chabi B., Wrutniak-Cabello C., Tong Q. Regulation of Skeletal Muscle Oxidative Capacity and Muscle Mass by SIRT3. PLoS ONE. 2014;9:e85636. doi: 10.1371/journal.pone.0085636.
    1. Sundaresan N.R., Gupta M., Kim G., Rajamohan S.B., Isbatan A., Gupta M.P. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J. Clin. Investig. 2009;119:2758–2771. doi: 10.1172/JCI39162.
    1. Ma Y., Chai H., Ding Q., Qian Q., Yan Z., Ding B., Dou X., Li S. Hepatic SIRT3 Upregulation in Response to Chronic Alcohol Consumption Contributes to Alcoholic Liver Disease in Mice. Front. Physiol. 2019;10:1042. doi: 10.3389/fphys.2019.01042.
    1. Lombard D.B., Zwaans B.M.M. SIRT3: As Simple As It Seems? Gerontology. 2014;60:56–64. doi: 10.1159/000354382.
    1. Hirschey M.D., Shimazu T., Goetzman E., Jing E., Schwer B., Lombard D.B., Grueter C.A., Harris C., Biddinger S., Ilkayeva O.R., et al. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature. 2010;464:121–125. doi: 10.1038/nature08778.
    1. Shimazu T., Hirschey M.D., Hua L., Dittenhafer-Reed K.E., Schwer B., Lombard D.B., Li Y., Bunkenborg J., Alt F.W., Denu J.M., et al. SIRT3 Deacetylates Mitochondrial 3-Hydroxy-3-Methylglutaryl CoA Synthase 2 and Regulates Ketone Body Production. Cell Metab. 2010;12:654–661. doi: 10.1016/j.cmet.2010.11.003.
    1. Liu L., Nam M., Fan W., Akie T.E., Hoaglin D.C., Gao G., Keaney J.F., Jr., Cooper M.P. Nutrient sensing by the mitochondrial transcription machinery dictates oxidative phosphorylation. J. Clin. Investig. 2014;124:768–784. doi: 10.1172/JCI69413.
    1. Jing E., O’Neill B.T., Rardin M.J., Kleinridders A., Ilkeyeva O.R., Ussar S., Bain J.R., Lee K.Y., Verdin E.M., Newgard C.B., et al. Sirt3 Regulates Metabolic Flexibility of Skeletal Muscle Through Reversible Enzymatic Deacetylation. Diabetes. 2013;62:3404–3417. doi: 10.2337/db12-1650.
    1. Li M., Li C.-M., Ye Z.-C., Huang J., Li Y., Lai W., Peng H., Lou T.-Q. Sirt3 modulates fatty acid oxidation and attenuates cisplatin-induced AKI in mice. J. Cell. Mol. Med. 2020;24:5109–5121. doi: 10.1111/jcmm.15148.
    1. Ye X., Li M., Hou T., Gao T., Zhu W.-g., Yang Y. Sirtuins in glucose and lipid metabolism. Oncotarget. 2017;8:1845–1859. doi: 10.18632/oncotarget.12157.
    1. Cheung K.G., Cole L.K., Xiang B., Chen K., Ma X., Myal Y., Hatch G.M., Tong Q., Dolinsky V.W. Sirtuin-3 (SIRT3) Protein Attenuates Doxorubicin-induced Oxidative Stress and Improves Mitochondrial Respiration in H9c2 Cardiomyocytes. J. Biol. Chem. 2015;290:10981–10993. doi: 10.1074/jbc.M114.607960.
    1. Jiang X., Zhang Y., Hu W., Liang Y., Zheng L., Zheng J., Wang B., Guo X. Different Effects of Leucine Supplementation and/or Exercise on Systemic Insulin Sensitivity in Mice. Front. Endocrinol. 2021;12:651303. doi: 10.3389/fendo.2021.651303.
    1. Guo X., Zhu B., Xu H., Li H., Jiang B., Wang Y., Zheng B., Glaser S., Alpini G., Wu C. Adoptive transfer of Pfkfb3-disrupted hematopoietic cells to wild-type mice exacerbates diet-induced hepatic steatosis and inflammation. Liver Res. 2020;4:136–144. doi: 10.1016/j.livres.2020.08.004.
    1. Zhu B., Guo X., Xu H., Jiang B., Li H., Wang Y., Yin Q., Zhou T., Cai J.J., Glaser S., et al. Adipose tissue inflammation and systemic insulin resistance in mice with diet-induced obesity is possibly associated with disruption of PFKFB3 in hematopoietic cells. Lab. Investig. 2021;101:328–340. doi: 10.1038/s41374-020-00523-z.
    1. Guo X., Shu C., Li H., Pei Y., Woo S.-L., Zheng J., Liu M., Xu H., Botchlett R., Guo T., et al. Cyclic GMP-AMP Ameliorates Diet-induced Metabolic Dysregulation and Regulates Proinflammatory Responses Distinctly from STING Activation. Sci. Rep. 2017;7:6355. doi: 10.1038/s41598-017-05884-y.
    1. Ferchaud-Roucher V., Barner K., Jansson T., Powell T.L. Maternal obesity results in decreased syncytiotrophoblast synthesis of palmitoleic acid, a fatty acid with anti-inflammatory and insulin-sensitizing properties. FASEB J. 2019;33:6643–6654. doi: 10.1096/fj.201802444R.
    1. Frigolet M.E., Gutierrez-Aguilar R. The Role of the Novel Lipokine Palmitoleic Acid in Health and Disease. Adv. Nutr. 2017;8:173S–181S. doi: 10.3945/an.115.011130.
    1. Liu Y., Shen X., Pang M., Sun Z., Qian Y., Xue W., Wang Z., Li L. Role of histone deacetylase Sirt3 in the development and regression of atherosclerosis. Life Sci. 2021;272:119178. doi: 10.1016/j.lfs.2021.119178.
    1. Giralt A., Villarroya F. SIRT3, a pivotal actor in mitochondrial functions: Metabolism, cell death and aging. Biochem. J. 2012;444:1–10. doi: 10.1042/BJ20120030.
    1. Nakamura A., Kawakami K., Kametani F., Goto S. Dietary Restriction Increases Protein Acetylation in the Livers of Aged Rats. Gerontology. 2013;59:542–548. doi: 10.1159/000354087.
    1. Hebert A.S., Dittenhafer-Reed K.E., Yu W., Bailey D.J., Selen E.S., Boersma M.D., Carson J.J., Tonelli M., Balloon A.J., Higbee A.J., et al. Calorie Restriction and SIRT3 Trigger Global Reprogramming of the Mitochondrial Protein Acetylome. Mol. Cell. 2013;49:186–199. doi: 10.1016/j.molcel.2012.10.024.
    1. Arima Y., Nakagawa Y., Takeo T., Ishida T., Yamada T., Hino S., Nakao M., Hanada S., Umemoto T., Suda T., et al. Murine neonatal ketogenesis preserves mitochondrial energetics by preventing protein hyperacetylation. Nat. Metab. 2021;3:196–210. doi: 10.1038/s42255-021-00342-6.
    1. Nie Y.Z., Erion D.M., Yuan Z.L., Dietrich M., Shulman G.I., Horvath T.L., Gao Q. STAT3 inhibition of gluconeogenesis is downregulated by SirT1. Nat. Cell Biol. 2009;11:492–500. doi: 10.1038/ncb1857.
    1. Petersen M.C., Vatner D.F., Shulman G.I. Regulation of hepatic glucose metabolism in health and disease. Nat. Rev. Endocrinol. 2017;13:572–587. doi: 10.1038/nrendo.2017.80.
    1. Rui L. Energy Metabolism in the Liver. Compr. Physiol. 2014;4:177–197. doi: 10.1002/cphy.c130024.
    1. Narita T., Weinert B.T., Choudhary C. Functions and mechanisms of non-histone protein acetylation. Nat. Rev. Mol. Cell Biol. 2019;20:156–174. doi: 10.1038/s41580-018-0081-3.
    1. De Wit N., Derrien M., Bosch-Vermeulen H., Oosterink E., Keshtkar S., Duval C., de Vogel-van den Bosch J., Kleerebezem M., Muller M., van der Meer R. Saturated fat stimulates obesity and hepatic steatosis and affects gut microbiota composition by an enhanced overflow of dietary fat to the distal intestine. Am. J. Physiol.-Gastrointest. Liver Physiol. 2012;303:G589–G599. doi: 10.1152/ajpgi.00488.2011.
    1. Buettner R., Parhofer K.G., Woenckhaus M., Wrede C.E., Kunz-Schughart L.A., Schoelmerich J., Bollheimer L.C. Defining high-fat-diet rat models: Metabolic and molecular effects of different fat types. J. Mol. Endocrinol. 2006;36:485–501. doi: 10.1677/jme.1.01909.
    1. Zbikowski A., Blachnio-Zabielska A., Galli M., Zabielski P. Adipose-Derived Exosomes as Possible Players in the Development of Insulin Resistance. Int. J. Mol. Sci. 2021;22:7427. doi: 10.3390/ijms22147427.
    1. Al-Mansoori L., Al-Jaber H., Prince M.S., Elrayess M.A. Role of Inflammatory Cytokines, Growth Factors and Adipokines in Adipogenesis and Insulin Resistance. Inflammation. 2022;45:31–44. doi: 10.1007/s10753-021-01559-z.
    1. Nunes E.A., Rafacho A. Implications of Palmitoleic Acid (Palmitoleate) On Glucose Homeostasis, Insulin Resistance and Diabetes. Curr. Drug Targets. 2017;18:619–628. doi: 10.2174/1389450117666151209120345.
    1. Souza C.O., Teixeira A.A.S., Lima E.A., Batatinha H.A.P., Gomes L.M., Carvalho-Silva M., Mota I.T., Streck E.L., Hirabara S.M., Rosa Neto J.C. Palmitoleic Acid (N-7) Attenuates the Immunometabolic Disturbances Caused by a High-Fat Diet Independently of PPAR alpha. Mediat. Inflamm. 2014;2014:582197. doi: 10.1155/2014/582197.
    1. Cao H.M., Gerhold K., Mayers J.R., Wiest M.M., Watkins S.M., Hotamisligil G.S. Identification of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism. Cell. 2008;134:933–944. doi: 10.1016/j.cell.2008.07.048.
    1. Pinnick K.E., Neville M.J., Fielding B.A., Frayn K.N., Karpe F., Hodson L. Gluteofemoral Adipose Tissue Plays a Major Role in Production of the Lipokine Palmitoleate in Humans. Diabetes. 2012;61:1399–1403. doi: 10.2337/db11-1810.
    1. Stefan N., Kantartzis K., Celebi N., Staiger H., Machann J., Schick F., Cegan A., Elcnerova M., Schleicher E., Fritsche A., et al. Circulating Palmitoleate Strongly and Independently Predicts Insulin Sensitivity in Humans. Diabetes Care. 2010;33:405–407. doi: 10.2337/dc09-0544.
    1. De Souza C.O., Vannice G.K., Rosa Neto J.C., Calder P.C. Is Palmitoleic Acid a Plausible Nonpharmacological Strategy to Prevent or Control Chronic Metabolic and Inflammatory Disorders? Mol. Nutr. Food Res. 2018;62:1700504. doi: 10.1002/mnfr.201700504.
    1. Campbell J.E., Newgard C.B. Mechanisms controlling pancreatic islet cell function in insulin secretion. Nat. Rev. Mol. Cell Biol. 2021;22:142–158. doi: 10.1038/s41580-020-00317-7.
    1. Hutfles L.J., Wilkins H.M., Koppel S.J., Weidling I.W., Selfridge J.E., Tan E., Thyfault J.P., Slawson C., Fenton A.W., Zhu H., et al. A bioenergetics systems evaluation of ketogenic diet liver effects. Appl. Physiol. Nutr. Metab. 2017;42:955–962. doi: 10.1139/apnm-2017-0068.
    1. Wang Q., Zhang Y., Yang C., Xiong H., Lin Y., Yao J., Li H., Xie L., Zhao W., Yao Y., et al. Acetylation of Metabolic Enzymes Coordinates Carbon Source Utilization and Metabolic Flux. Science. 2010;327:1004–1007. doi: 10.1126/science.1179687.
    1. Yang H., Zhou L., Shi Q., Zhao Y., Lin H., Zhang M., Zhao S., Yang Y., Ling Z.-Q., Guan K.-L., et al. SIRT3-dependent GOT2 acetylation status affects the malate-aspartate NADH shuttle activity and pancreatic tumor growth. EMBO J. 2015;34:1110–1125. doi: 10.15252/embj.201591041.
    1. Burgess S.C., He T., Yan Z., Lindner J., Sherry A.D., Malloy C.R., Browning J.D., Magnuson M.A. Cytosolic phosphoenolpyruvate carboxykinase does not solely control the rate of hepatic gluconeogenesis in the intact mouse liver. Cell Metab. 2007;5:313–320. doi: 10.1016/j.cmet.2007.03.004.
    1. Jiang W., Wang S., Xiao M., Lin Y., Zhou L., Lei Q., Xiong Y., Guan K.-L., Zhao S. Acetylation Regulates Gluconeogenesis by Promoting PEPCK1 Degradation via Recruiting the UBR5 Ubiquitin Ligase. Mol. Cell. 2011;43:33–44. doi: 10.1016/j.molcel.2011.04.028.
    1. Latorre-Muro P., Baeza J., Armstrong E.A., Hurtado-Guerrero R., Corzana F., Wu L.E., Sinclair D.A., Lopez-Buesa P., Carrodeguas J.A., Denu J.M. Dynamic Acetylation of Phosphoenolpyruvate Carboxykinase Toggles Enzyme Activity between Gluconeogenic and Anaplerotic Reactions. Mol. Cell. 2018;71:718–732. doi: 10.1016/j.molcel.2018.07.031.

Source: PubMed

3
Suscribir