Journal Home > Volume 13 , Issue 2

With the prevalence of obesity and obesity-related metabolic syndrome, such as insulin resistance in recent years, it is urgent to explore effective interventions to prevent the progression of obesity-related metabolic syndrome. Palmitoleic acid is a monounsaturated fatty acid that is available from dietary sources, mainly derived from marine products. Palmitoleic acid plays a positive role in maintaining glucose homeostasis and reducing inflammation. However, it is still unknow the mechanism of palmitoleic acid in ameliorating insulin resistance. Here, we investigated the effects of palmitoleic acid on chow diet (CD)-fed and high-fat diet (HFD)-fed mice, which were fed CD or HFD for 12 weeks before administration. We administrated mice with BSA (control), oleic acid, or palmitoleic acid for 6 weeks on top of CD or HFD feeding. We found that palmitoleic acid only improved glucose homeostasis in HFD-fed obese mice by increasing glucose clearance and reducing HOMA-IR. Further study explored that palmitoleic acid changed the composition of gut microbiota by decreasing Firmicutes population and increasing Bacteroidetes population. In colon, palmitoleic acid increased intestinal tight junction integrity and reduced inflammation. Moreover, palmitoleic acid decreased macrophage infiltration in liver and adipose tissue and increase glucose uptake in adipose tissue. Diacylglycerol (DAG) in tissue (for example, liver) is found to positively correlated with HOMA-IR. HFD enhanced the levels of DAGs in liver but not in adipose tissue in this study. Palmitoleic acid did not reverse the high DAG levels induced by HFD in liver. Therefore, in HFD-fed mice, palmitoleic acid reduced insulin resistance by an independent-manner of DAGs. It might be associated with the beneficial effects of palmitoleic acid on altering the gut microbiota composition, improving of intestinal barrier function, and downregulating the inflammation in colon, liver, and adipose tissue.


menu
Abstract
Full text
Outline
Electronic supplementary material
About this article

Palmitoleic acid on top of HFD ameliorates insulin resistance independent of diacylglycerols and alters gut microbiota in C57BL/6J mice

Show Author's information Qijian LiangaYan ZhengbFanli Mengc,dXiaofan JiangaQingcai ZhenaZhongting LuaShixiu ZhangaLei DuaHao Wua( )Xin Guoa,( )
Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
Research Center of Translational Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
Institute of Hepatology, Shandong University, Jinan 250012, China

Peer review under responsibility of Tsinghua University Press.

Highlights

• Palmitoleic acid alleviates HFD-induced insulin resistance.

• Palmitoleic acid decreased Firmicutes population and increased Bacteroidetes population in gut of HFD-fed mice.

• Palmitoleic acid increased tight junction integrity and reduced inflammation in colon.

• Palmitoleic acid decreased inflammation in liver and adipose tissue.

Abstract

With the prevalence of obesity and obesity-related metabolic syndrome, such as insulin resistance in recent years, it is urgent to explore effective interventions to prevent the progression of obesity-related metabolic syndrome. Palmitoleic acid is a monounsaturated fatty acid that is available from dietary sources, mainly derived from marine products. Palmitoleic acid plays a positive role in maintaining glucose homeostasis and reducing inflammation. However, it is still unknow the mechanism of palmitoleic acid in ameliorating insulin resistance. Here, we investigated the effects of palmitoleic acid on chow diet (CD)-fed and high-fat diet (HFD)-fed mice, which were fed CD or HFD for 12 weeks before administration. We administrated mice with BSA (control), oleic acid, or palmitoleic acid for 6 weeks on top of CD or HFD feeding. We found that palmitoleic acid only improved glucose homeostasis in HFD-fed obese mice by increasing glucose clearance and reducing HOMA-IR. Further study explored that palmitoleic acid changed the composition of gut microbiota by decreasing Firmicutes population and increasing Bacteroidetes population. In colon, palmitoleic acid increased intestinal tight junction integrity and reduced inflammation. Moreover, palmitoleic acid decreased macrophage infiltration in liver and adipose tissue and increase glucose uptake in adipose tissue. Diacylglycerol (DAG) in tissue (for example, liver) is found to positively correlated with HOMA-IR. HFD enhanced the levels of DAGs in liver but not in adipose tissue in this study. Palmitoleic acid did not reverse the high DAG levels induced by HFD in liver. Therefore, in HFD-fed mice, palmitoleic acid reduced insulin resistance by an independent-manner of DAGs. It might be associated with the beneficial effects of palmitoleic acid on altering the gut microbiota composition, improving of intestinal barrier function, and downregulating the inflammation in colon, liver, and adipose tissue.

Keywords: Gut microbiota, Insulin resistance, High fat diet, Palmitoleic acid

References(80)

[1]

M. Blüher, Obesity: global epidemiology and pathogenesis, Nat. Rev. Endocrinol. 15(5) (2019) 288-298. https://doi.org/10.1038/s41574-019-0176-8.

[2]

R. Barazzoni, G.G. Cappellari, M. Ragni, et al., Insulin resistance in obesity: an overview of fundamental alterations, Eat. Weight Disord. 23(2) (2018) 149-157. https://doi.org/10.1007/s40519-018-0481-6.

[3]

H. Wu, C.M. Ballantyne, Metabolic inflammation and insulin resistance in obesity, Circul. Res. 126(11) (2020) 1549-1564. https://doi.org/10.1161/circresaha.119.315896.

[4]

B. Zhu, X. Guo, H. Xu, 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. Invest. 101(3) (2021) 328-340. https://doi.org/10.1038/s41374-020-00523-z.

[5]

F. Zatterale, M. Longo, J. Naderi, et al., Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes, Front. Physiol. 10 (2020). https://doi.org/10.3389/fphys.2019.01607.

[6]

F. Bessone, M. Valeria Razori, M.G. Roma, Molecular pathways of nonalcoholic fatty liver disease development and progression, Cell. Mol. Life Sci. 76(1) (2019) 99-128. https://doi.org/10.1007/s00018-018-2947-0.

[7]

K. Kazankov, S.M.D. Jorgensen, K.L. Thomsen, et al., The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, Nat. Rev. Gastroenterol. Hepatol. 16(3) (2019) 145-159. https://doi.org/10.1038/s41575-018-0082-x.

[8]

S.M. Wright, L.J. Aronne, Causes of obesity, Abdom. Imaging. 37(5) (2012) 730-732. https://doi.org/10.1007/s00261-012-9862-x.

[9]

J.R. Speakman, Use of high-fat diets to study rodent obesity as a model of human obesity, Int. J. Obes. (Lond.). 43(8) (2019) 1491-1492. https://doi.org/10.1038/s41366-019-0363-7.

[10]

A.L. Mundy, E. Haas, I. Bhattacharya, et al., Fat intake modifies vascular responsiveness and receptor expression of vasoconstrictors: implications for diet-induced obesity, Cardiovasc. Res. 73(2) (2007) 368-375. https://doi.org/10.1016/j.cardiores.2006.11.019.

[11]

S.E. Arnold, I. Lucki, B.R. Brookshire, et al., High fat diet produces brain insulin resistance, synaptodendritic abnormalities and altered behavior in mice, Neurobiol. Dis. 67 (2014) 79-87. https://doi.org/10.1016/j.nbd.2014.03.011.

[12]

K. Gil-Cardoso, I. Gines, M. Pinent, et al., Effects of flavonoids on intestinal inflammation, barrier integrity and changes in gut microbiota during diet-induced obesity, Nutr. Res. Rev. 29(2) (2016) 234-248. https://doi.org/10.1017/s0954422416000159.

[13]

S. Ding, P.K. Lund, Role of intestinal inflammation as an early event in obesity and insulin resistance, Curr. Opin. Clin. Nutr. Metab. Care. 14(4) (2011) 328-333. https://doi.org/10.1097/MCO.0b013e3283478727.

[14]

C. Torres-Fuentes, H. Schellekens, T.G. Dinan, et al., The microbiota-gutbrain axis in obesity, Lancet Gastroenterol. Hepatol. 2(10) (2017) 747-756. https://doi.org/10.1016/s2468-1253(17)30147-4.

[15]

V. Tremaroli, F. Karlsson, M. Werling, et al., Roux-en-Y gastric bypass and vertical banded gastroplasty induce long-term changes on the human gut microbiome contributing to fat mass regulation, Cell Metab. 22(2) (2015) 228-238. https://doi.org/10.1016/j.cmet.2015.07.009.

[16]

Z. Zhang, V. Mocanu, C. Cai, et al., Impact of fecal microbiota transplantation on obesity and metabolic syndrome-a systematic review, Nutrients 11(10) (2019). https://doi.org/10.3390/nu11102291.

[17]

C. Barcena, R. Valdes-Mas, P. Mayoral, et al, Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice, Nat. Med. 25(8) (2019) 1234-1242. https://doi.org/10.1038/s41591-019-0504-5.

[18]

J.R. Allegretti, Z. Kassam, B.H. Mullish, et al, Effects of fecal microbiota transplantation with oral capsules in obese patients, Clin. Gastroenterol. Hepatol. 18(4) (2020) 855-863. https://doi.org/10.1016/j.cgh.2019.07.006.

[19]

Y.C. Orbe-Orihuela, A. Lagunas-Martinez, M. Bahena-Roman, et al, High relative abundance of firmicutes and increased TNF-alpha levels correlate with obesity in children, Salud. Publica. Mex. 60(1) (2018) 5-11. https://doi.org/10.21149/8133.

[20]

A. Pascale, N. Marchesi, S. Govoni, et al., The role of gut microbiota in obesity, diabetes mellitus, and effect of metformin: new insights into old diseases, Curr. Opin. Pharm. 49 (2019) 1-5. https://doi.org/10.1016/j.coph.2019.03.011.

[21]

J. Zhang, Y. Ni, L. Qian, et al, Decreased abundance of Akkermansia muciniphila leads to the impairment of insulin secretion and glucose homeostasis in lean type 2 diabetes, Adv. Sci. 8(16) (2021). https://doi.org/10.1002/advs.202100536.

[22]

C. Depommier, A. Everard, C. Druart, et al, Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study, Nat. Med. 25(7) (2019) 1096-1103. https://doi.org/10.1038/s41591-019-0495-2.

[23]

X. Jiang, J. Zheng, S. Zhang, et al., Advances in the involvement of gut microbiota in pathophysiology of NAFLD, Front. Med. 7 (2020). https://doi.org/10.3389/fmed.2020.00361.

[24]

Y.Y. Lam, C.W. Ha, J.M. Hoffmann, et al., Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice, Obesity 23(7) (2015) 1429-1439. https://doi.org/10.1002/oby.21122.

[25]

R. Caesar, V. Tremaroli, P. Kovatcheva-Datchary, et al., Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signaling, Cell Metab. 22(4) (2015) 658-668. https://doi.org/10.1016/j.cmet.2015.07.026.

[26]

N. de Wit, M. Derrien, H. Bosch-Vermeulen, et al., 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. 303(5) (2012) G589-599. https://doi.org/10.1152/ajpgi.00488.2011.

[27]

E. Patterson, O.D. RM, E.F. Murphy, et al., Impact of dietary fatty acids on metabolic activity and host intestinal microbiota composition in C57BL/6J mice, Br. J. Nutr. 111(11) (2014) 1905-1917. https://doi.org/10.1017/s0007114514000117.

[28]

J.A. Jackman, B.K. Yoon, D. Li, et al., Nanotechnology formulations for antibacterial free fatty acids and monoglycerides, Molecules 21(3) (2016) 305. https://doi.org/10.3390/molecules21030305.

[29]

J. Ogawa, S. Kishino, A. Ando, et al., Production of conjugated fatty acids by lactic acid bacteria, J. Biosci. Bioeng. 100(4) (2005) 355-364. https://doi.org/10.1263/jbb.100.355.

[30]

M. Schoeler, R. Caesar, Dietary lipids, gut microbiota and lipid metabolism, Rev. Endocr. Metab. Disord. 20(4) (2019) 461-472. https://doi.org/10.1007/s11154-019-09512-0.

[31]

M.E. Frigolet, R. Gutiérrez-Aguilar, The role of the novel lipokine palmitoleic acid in health and disease, Adv. Nutr. 8(1) (2017) 173s-181s. https://doi.org/10.3945/an.115.011130.

[32]

E.A. Nunes, A. Rafacho, Implications of palmitoleic acid (Palmitoleate) on glucose homeostasis, insulin resistance and diabetes, Curr. Drug Targets. 18(6) (2017) 619-628. https://doi.org/10.2174/1389450117666151209120345.

[33]

C.O. Souza, A.A.S. Teixeira, L.A. Biondo, et al, Palmitoleic acid reduces high fat diet-induced liver inflammation by promoting PPAR-γ-independent M2a polarization of myeloid cells, Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 1865(10) (2020) 158776. https://doi.org/10.1016/j.bbalip.2020.158776.

[34]

Z.H. Yang, M. Pryor, A. Noguchi, et al., Dietary palmitoleic acid attenuates atherosclerosis progression and hyperlipidemia in low-density lipoprotein receptor-deficient mice, Mol. Nutr. Food Res. 63(12) (2019) e1900120. https://doi.org/10.1002/mnfr.201900120.

[35]

I. Çimen, B. Kocatürk, S. Koyuncu, et al, Prevention of atherosclerosis by bioactive palmitoleate through suppression of organelle stress and inflammasome activation, Sci. Transl. Med. 8(358) (2016) 358ra126. https://doi.org/10.1126/scitranslmed.aaf9087.

[36]

A. Bolsoni-Lopes, W.T. Festuccia, P. Chimin, et al., Palmitoleic acid (n-7) increases white adipocytes GLUT4 content and glucose uptake in association with AMPK activation, Lipids Health Dis. 13 (2014) 199. https://doi.org/10.1186/1476-511x-13-199.

[37]

N.A. Talbot, C.P. Wheeler-Jones, M.E. Cleasby, Palmitoleic acid prevents palmitic acid-induced macrophage activation and consequent p38 MAPKmediated skeletal muscle insulin resistance, Mol. Cell. Endocrinol. 393(1/2) (2014) 129-142. https://doi.org/10.1016/j.mce.2014.06.010.

[38]

H.J. Welters, E. Diakogiannaki, J.M. Mordue, et al., Differential protective effects of palmitoleic acid and cAMP on caspase activation and cell viability in pancreatic beta-cells exposed to palmitate, Apoptosis 11(7) (2006) 12311238. https://doi.org/10.1007/s10495-006-7450-7.

[39]

G. Singh, A. Brass, S.M. Cruickshank, et al., Cage and maternal effects on the bacterial communities of the murine gut, Sci. Rep. 11(1) (2021). https://doi.org/10.1038/s41598-021-89185-5.

[40]

I. Bakoyiannis, Reducing cage effects in mouse microbiome studies, Lab Animal 51(7) (2022) 185. https://doi.org/10.1038/s41684-022-01009-9.

[41]

T. Magoč, S.L. Salzberg, FLASH: fast length adjustment of short reads to improve genome assemblies, Bioinformatics 27(21) (2011) 2957-2963. https://doi.org/10.1093/bioinformatics/btr507

[42]

N.A. Bokulich, S. Subramanian, J.J. Faith, et al., Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing, Nat. Methods 10(1) (2013) 57-59. https://doi.org/10.1038/nmeth.2276.

[43]

J.G. Caporaso, J. Kuczynski, J. Stombaugh, et al, QIIME allows analysis of high-throughput community sequencing data, Nat. Methods 7(5) (2010) 335336. https://doi.org/10.1038/nmeth.f.303.

[44]

R.C. Edgar, UPARSE: highly accurate OTU sequences from microbial amplicon reads, Nat. Methods 10(10) (2013) 996-998. https://doi.org/10.1038/nmeth.2604.

[45]

C. Quast, E. Pruesse, P. Yilmaz, et al., The SILVA ribosomal RNA gene database project: improved data processing and web-based tools, Nucleic Acids Res. 41 (2013) D590-596. https://oi.org/10.1093/nar/gks1219.

[46]

R.C. Edgar, MUSCLE: multiple sequence alignment with high accuracy and high throughput, Nucleic Acids Res. 32(5) (2004) 1792-1797. https://doi.org/10.1093/nar/gkh340.

[47]

X. Jiang, Y. Zhang, W. Hu, Y. Liang, et al., Different effects of leucine supplementation and/or exercise on systemic insulin sensitivity in mice, Front. Endocrinol. (Lausanne). 12 (2021) 651303. https://doi.org/10.3389/fendo.2021.651303.

[48]

R. Barazzoni, G. Gortan Cappellari, M. Ragni, et al., Insulin resistance in obesity: an overview of fundamental alterations, Eat. Weight Disord. 23(2) (2018) 149-157. https://doi.org/10.1007/s40519-018-0481-6.

[49]

X. Guo, H. Li, H. Xu, et al., Disruption of inducible 6-phosphofructo-2kinase impairs the suppressive effect of PPAR γ activation on diet-induced intestine inflammatory response, J. Nutr. Biochem. 24(5) (2013) 770-775. https://doi.org/10.1016/j.jnutbio.2012.04.007.

[50]

L. Du, Y.M. Hao, Y.H. Yang, et al., DHA-enriched phospholipids and EPAenriched phospholipids alleviate lipopolysaccharide-induced intestinal barrier injury in mice via a sirtuin 1-dependent mechanism, J. Agric. Food Chem. 70(9) (2022) 2911-2922. https://doi.org/10.1021/acs.jafc.1c07761.

[51]

Y. Huo, X. Guo, H. Li, et al., Disruption of inducible 6-phosphofructo-2kinase ameliorates diet-induced adiposity but exacerbates systemic insulin resistance and adipose tissue inflammatory response, J. Biol. Chem. 285(6) (2010) 3713-3721. https://doi.org/10.1074/jbc.M109.058446.

[52]

X. Guo, B. Zhu, H. Xu, et al., Adoptive transfer of Pfkfb3-disrupted hematopoietic cells to wild-type mice exacerbates diet-induced hepatic steatosis and inflammation, Liver Res. 4(3) (2020) 136-144. https://doi.org/10.1016/j.livres.2020.08.004.

[53]

M.C. Petersen, G.I. Shulman, Roles of diacylglycerols and ceramides in hepatic insulin resistance, Trends Pharmacol. Sci. 38(7) (2017) 649-665. https://doi.org/10.1016/j.tips.2017.04.004.

[54]

F.R. Jornayvaz, G.I. Shulman, Diacylglycerol activation of protein kinase Cε and hepatic insulin resistance, Cell Metab. 15(5) (2012) 574-584. https://doi.org/10.1016/j.cmet.2012.03.005.

[55]

Y. Fan, O. Pedersen, Gut microbiota in human metabolic health and disease, Nat. Rev. Microbiol. 19(1) (2021) 55-71. https://doi.org/10.1038/s41579-020-0433-9.

[56]

M. Manco, L. Putignani, G.F. Bottazzo, Gut microbiota, lipopolysaccharides, and innate immunity in the pathogenesis of obesity and cardiovascular risk, Endocr. Rev. 31(6) (2010) 817-844. https://doi.org/10.1210/er.2009-0030.

[57]

L. Ma, Y. Ni, Z. Wang, W. Tu, et al., Spermidine improves gut barrier integrity and gut microbiota function in diet-induced obese mice, Gut Microbes. 12(1) (2020) 1-19. https://doi.org/10.1080/19490976.2020.1832857.

[58]

A. Kashani, A.D. Brejnrod, C. Jin, et al., Impaired glucose metabolism and altered gut microbiome despite calorie restriction of ob/ob mice, Anim. Microbiome. 1(1) (2019) 11. https://doi.org/10.1186/s42523-019-0007-1.

[59]

Y. Kang, Y. Cai, Gut microbiota and obesity: implications for fecal microbiota transplantation therapy, Hormones (Athens). 16(3) (2017) 223234. https://doi.org/10.14310/horm.2002.1742.

[60]

R.R. Rodrigues, R.L. Greer, X. Dong, et al., Antibiotic-induced alterations in gut microbiota are associated with changes in glucose metabolism in healthy mice, Front. Microbiol. 8 (2017) 2306. https://doi.org/10.3389/fmicb.2017.02306.

[61]

J.C. Aristizabal, L.I. González-Zapata, A. Estrada-Restrepo, et al., Concentrations of plasma free palmitoleic and dihomo-gamma linoleic fatty acids are higher in children with abdominal obesity, Nutrients 10(1) (2018). https://doi.org/10.3390/nu10010031.

[62]

X. Guo, H. Li, H. Xu, et al, Palmitoleate induces hepatic steatosis but suppresses liver inflammatory response in mice, PLoS One 7(6) (2012) e39286. https://doi.org/10.1371/journal.pone.0039286.

[63]

W. Zhang-Sun, L.A. Augusto, L. Zhao, et al., Desulfovibrio desulfuricans isolates from the gut of a single individual: structural and biological lipid A characterization, FEBS Lett. 589(1) (2015) 165-171. https://doi.org/10.1016/j.febslet.2014.11.042.

[64]

P. Paone, P.D. Cani, Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut 69(12) (2020) 2232-2243. https://doi.org/10.1136/gutjn1-2020-322260.

[65]

Z. Li, G. Quan, X. Jiang, et al., Effects of metabolites derived from gut microbiota and hosts on pathogens, Front. Cell Infect. Microbiol. 8 (2018) 314. https://doi.org/10.3389/fcimb.2018.00314.

[66]

S. Sivaprakasam, Y.D. Bhutia, S. Yang, et al., Short-chain fatty acid transporters: role in colonic homeostasis, Compr. Physiol. 8(1) (2017) 299314. https://doi.org/10.1002/cphy.c170014.

[67]

J.M. Natividad, A. Agus, J. Planchais, et al., Impaired aryl hydrocarbon receptor ligand production by the gut microbiota is a key factor in metabolic syndrome, Cell. Metab. 28(5) (2018) 737-749. https://doi.org/10.1016/j.cmet.2018.07.001.

[68]

B. Ey, A. Eyking, G. Gerken, et al., TLR2 mediates gap junctional intercellular communication through connexin-43 in intestinal epithelial barrier injury, J. Biol. Chem. 284(33) (2009) 22332-22343. https://doi.org/10.1074/jbc.M901619200.

[69]

C.A. Thaiss, M. Levy, I. Grosheva, et al, Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection, Science 359(6382) (2018) 1376-1383. https://doi.org/10.1126/science.aar3318.

[70]

J. Shi, L. Wang, Y. Lu, et al., Protective effects of seabuckthorn pulp and seed oils against radiation-induced acute intestinal injury, J. Radiat. Res. 58(1) (2017) 24-32. https://doi.org/10.1093/jrr/rrw069.

[71]

N. Bueno-Hernández, M.S. Sixtos-Alonso, M.D.P. Milke García, et al., Effect of cis-palmitoleic acid supplementation on inflammation and expression of HNF4γ, HNF4α and IL6 in patients with ulcerative colitis, Minerva Gastroenterol. Dietol. 63(3) (2017) 257-263. https://doi.org/10.23736/s1121-421x.17.02367-4.

[72]

S. Terzo, F. Mulè, G.F. Caldara, et al., Pistachio consumption alleviates inflammation and improves gut microbiota composition in mice fed a highfat diet, Int. J. Mol. Sci. 21(1) (2020). https://doi.org/10.3390/ijms21010365.

[73]

V. Kushwaha, P. Rai, S. Varshney, et al., Sodium butyrate reduces endoplasmic reticulum stress by modulating CHOP and empowers favorable anti-inflammatory adipose tissue immune-metabolism in HFD fed mice model of obesity, Food Chem. (Oxf). 4 (2022) 100079. https://doi.org/10.1016/j.fochms.2022.100079.

[74]

L.G. Hersoug, P. Møller, S. Loft, Role of microbiota-derived lipopolysaccharide in adipose tissue inflammation, adipocyte size and pyroptosis during obesity, Nutr. Res. Rev. 31(2) (2018) 153-163. https://doi.org/10.1017/s0954422417000269.

[75]

P. Portincasa, L. Bonfrate, M. Khalil, et a1., Intestinal barrier and permeability in health, obesity and NAFLD, Biomedicines 10(1) (2021). https://doi.org/10.3390/biomedicines10010083.

[76]

M. Cengiz, S. Ozenirler, S. Elbeg, Role of serum toll-like receptors 2 and 4 in non-alcoholic steatohepatitis and liver fibrosis, J. Gastroenterol. Hepatol. 30(7) (2015) 1190-1196. https://doi.org/10.1111/jgh.12924.

[77]

X. Meng, D. Zou, Z. Shi, et al., Dietary diacylglycerol prevents high-fat dietinduced lipid accumulation in rat liver and abdominal adipose tissue, Lipids 39(1) (2004) 37-41. https://doi.org/10.1007/s11745-004-1199-1.

[78]

M.P. Corcoran, S. Lamon-Fava, R.A. Fielding, Skeletal muscle lipid deposition and insulin resistance: effect of dietary fatty acids and exercise, Am. J. Clin. Nutr. 85(3) (2007) 662-677. https://doi.org/10.1093/ajcn/85.3.662.

[79]

E. Gart, K. Salic, M.C. Morrison, et al, The human milk oligosaccharide 2 '-fucosyllactose alleviates liver steatosis, ER stress and insulin resistance by reducing hepatic diacylglycerols and improved gut permeability in obese Ldlr-/-.Leiden mice, Front. Nutr. 9 (2022) 904740. https://doi.org/10.3389/fnut.2022.904740.

[80]

M. Chacińska, P. Zabielski, M. Książek, et al., The impact of OMEGA-3 fatty acids supplementation on insulin resistance and content of adipocytokines and biologically active lipids in adipose tissue of high-fat diet fed rats, Nutrients 11(4) (2019). https://doi.org/10.3390/nu11040835.

File
fshw-13-2-856_ESM.docx (499.8 KB)
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 30 June 2022
Revised: 01 August 2022
Accepted: 22 October 2022
Published: 25 September 2023
Issue date: March 2024

Copyright

© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

Acknowledgements

This research was funded,in whole or in part,by National Natural Science Foundation of China (81803224) and Young Scholars Program of Shandong University (2018WLJH33) to X.G.; National Natural Science Foundation of China (81973031) and Cheeloo Young Scholar Program of Shandong University (21320089963054) to H.W.; Young Scholars Program of Shandong University (2018WLJH34)and the Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology (LMDBKF-2019-05) to L.D.

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return