eISSN: 2449-8238
ISSN: 2392-1099
Clinical and Experimental Hepatology
Current issue Archive Manuscripts accepted About the journal Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2023
vol. 9
 
Share:
Share:
Original paper

Increased hepatic Akt phosphorylation alleviated glucose intolerance and improved liver function in leptin-deficient mice

Tomer Adar
1
,
Meir Mizrahi
1
,
Yoav Lichtenstein
1
,
Yehudit Shabat
1
,
Rizan Sakhnini
2
,
Lida Zolotarov
1
,
Naim Shehadeh
2
,
Yaron Ilan
1

1.
Department of Medicine, Hadassah Medical Center and Faculty of Medicine, Hebrew University, Jerusalem, Israel
2.
Rambam Medical Center, Haifa, Israel
Clin Exp HEPATOL 2023; 9, 2: 164-171
Online publish date: 2023/06/23
Article file
- Increased.pdf  [0.14 MB]
Get citation
 
PlumX metrics:
 

Introduction

Several insulin activities are regulated by phosphatidylinositol 3-kinase (PI3K). Akt (protein kinase B) is a serine-threonine kinase with a pleckstrin homology domain and is an effector in some downstream signaling pathways of PI3K [1-3]. Akt also has a role in the PI3K-Akt-mTOR axis and is involved in the upregulation of the insulin-signaling pathways essential for maintaining glucose metabolism [2, 4]; it promotes glucose transport and liver glycogen synthesis, thus reducing blood glucose levels [4].

The PI3K/Akt signaling pathway plays a role in metabolism, and its imbalance is associated with developing type 2 diabetes and obesity [5]. The insulin receptor substrate (IRS)-1/PI3K/Akt signaling pathway mediates olanzapine-induced hepatic insulin resistance [6]. Increased O-linked β-N-acetylglucosamine (O-GlcNAc) is associated with insulin resistance in muscle and adipocytes [7]. Due to its characteristics, the PI3K/AKT signaling pathway is required for normal metabolism, and its imbalance leads to the development of obesity and type 2 diabetes mellitus [5]. Damage of the PI3K/AKT pathway in various body tissues leads to obesity and type 2 diabetes as the result of insulin resistance, and in turn, insulin resistance exacerbates the PI3K/AKT pathway, forming a vicious circle [8].

Increased susceptibility of the diabetic myocardium to ischemia/reperfusion-induced apoptosis/dysfunction was associated with decreased Akt activation [9]. Akt phosphorylation in islet β cells and diabetic mice promoted β-cell apoptosis [10]. Pancreatic fibroblast growth factor 21 (FGF21) protects T2DM mice by inducing PI3K/Akt signaling-dependent insulin expression and secretion [11]. Akt regulates pancreatic β-cell growth and survival, and its activation reduces the loss of functional islet mass [12].

PI3K/Akt signaling in the liver also affects FGF21-induced insulin expression and secretion [13]. Akt is associated with obesity-related target organ damage [14]. Insulin and insulin-like growth factor-1 stimulate specific arteries and are disrupted by diet-induced obesity. Following a long-term high-fat diet (HFD), insulin was a more potent activator of Akt than insulin-like growth factor-1 in the aorta, whereas, in resistance arteries, insulin-like growth factor-1 was more potent than insulin. PI3K/Akt is involved in insulin or insulin growth factor (IGF)-1-induced proliferation of intestinal crypts isolated from obese humans [15]. PI3K/Akt/Rac-1 signaling is also involved in the anti-inflammatory effect of insulin. Insulin inhibits high glucose-induced activation of p38, NF-κB, and STAT1 transcriptional activity by activating Akt-Rac-1 signaling [16].

Glycosphingolipids (GSLs) are involved in the pathogenesis of insulin resistance and associated target organ injury [17]. The GM3 ganglioside of caveolin-rich adipocytes binds to the insulin receptor (IR). Following insulin treatment, it dissociates its complex with caveolin, and lowers IR autophosphorylation [18]. The depletion of GSL in HepG2 cells affects both Akt1 phosphorylation and IR autophosphorylation by modifying the membrane microenvironment of this kinase [18].

Human hepatocyte-derived HepG2 cells are characterized by a high level of IR but a low level of caveolin. The glucosylceramide synthase inhibitors d-threo1-pheny-2-decanoylamino-3-morpholino-1-propanol (d-PDMP) and d-threo-1-(3,4,-ethylenedioxy)phenyl-2-palmitoylamino-3-pyrrolidino-1-propanol (d-EtDO-P4) lower the GM3 content, which was associated with a marked increase in Akt1 phosphorylation, and an increase in IR autophosphorylation following cell stimulation with insulin [18].

β-glucosylceramide (GC) is a GSL shown to exert immunomodulatory effects [19-27]. Oral administration of GC alleviated insulin resistance in several animal models [20, 21, 28], and preliminary results suggest a beneficial effect on type 2 diabetes and non-alcoholic fatty liver disease (NAFLD) [29].

The present study aimed to determine the effects of insulin and GC oral administration on Akt expression and metabolic syndrome characteristics in leptin-deficient ob/ob mice. Leptin regulates the long-term balance between the body’s food intake and energy expenditure [30]. Leptin-deficient ob/ob mice serve as a model of obesity-related liver steatosis and glucose intolerance [31].

Material and methods

Animals

Eight-week-old male ob/ob leptin-deficient mice, a model for obesity-associated liver steatosis and hyperglycemia [31], were purchased from Jackson Laboratories (Bar Harbor, ME). Mice were kept in the Animal Core of the Hadassah-Hebrew University Medical School. All mice were administered standard laboratory chow and water ad libitum and kept in a 12-hour light/dark cycle and at 24 degrees Celsius. The animal experiments were carried out under the supervision of the Hebrew University-Hadassah Institutional Committee for Care and Use of Laboratory Animals and with the committee’s approval.

Preparation of glycolipids

β-glucosylceramide was purchased from Avanti Polar Lipids (Alabaster, AL, USA), dissolved in a vehicle of a mixture of 30% Cremophor EL (Sigma, Rehovot,Israel) and ethanol (1 : 1) in PBS, and administered orally by gavage using a designated pipette at a dose of 2.5 mg/kg/day as described [21, 23, 25, 31].

Experimental groups

Four groups of leptin-deficient ob/ob mice (n = 6 each group) were orally administered by gavage using a designated pipette daily for four weeks: vehicle (group A); 2.5 mg/kg GC (group B); 2.5 IU regular insulin (Humulin R, Eli Lilly, group C); or a combination of GC with insulin (group D). The number of animals was calculated based on previously published data [21, 31]. Mice in all groups were followed for changes in metabolic parameters and parameters indicative of liver damage, as described [21, 23, 25]. The last treatment was administered the day before the end of the study. All studies were conducted in the morning hours.

Glucose tolerance test

The glucose tolerance test (GTT) was performed by oral administration of glucose (1.25 g/kg) at the end of the treatment period following a 6-hour fast, with repeated glucose level tests from the tail vein using a standard glucometer [31].

Liver enzymes

Serum alanine aminotransferase (ALT) was measured weekly using commercially available sticks (Reflovet Plus, Roche Diagnostic).

Triglyceride and total cholesterol levels

Triglyceride (TG) and total cholesterol serum levels were measured weekly in a fasted state using commercially available sticks with a Reflovet Plus clinical chemistry analyzer (Roche Diagnostics, GmbH, Mannheim, Germany).

Hepatic TG content

Intrahepatic TGs were quantified at the end of the study using a modification of the Folch method [32]. Quantification was followed by extracting by spectrophotometer, using a GPO-Trinder kit (Sigma, Rehovot, Israel), and levels were normalized to the protein content in the homogenate.

Serum cytokine levels

Tumor necrosis factor α serum levels were measured using a commercially available “sandwich” ELISA kit (Quantikine, R&D Systems, MN, USA) at the end of the treatment period.

Akt phosphorylation

Hepatic Akt phosphorylation was determined at the end of treatment at basal states by western blot analysis as described previously [33]. Whole tissue lysates from snap-frozen liver pieces were homogenized in modified RIPA buffer (Sigma). Lysates were cleared, and equivalent amounts of protein were used as determined by the Bradford (Bio-Rad) assay, separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Proteins were detected with phospho-specific Akt (Cell Signaling, USA, #9271); β-actin (Abcam, UK, sc-47778) was used as a loading control. All membranes were developed with horseradish peroxidase-conjugated secondary antibodies (Jackson Laboratories, USA) and Western Blot-Luminol Reagent (Santa Cruz Biotechnology, USA). Western blot films were scanned and quantified using Total Lab (Phoenix, USA) software.

Statistical analysis

Statistical analysis was performed using the ANOVAtest.

Results

Orally administered insulin and GC alleviated glucose intolerance and its accompanying liver injury. No significant changes in body weights were noted in all groups in the study.

Oral insulin and GC promoted hepatic Akt phosphorylation

Oral administration of GC and insulin, particularly their combination, was associated with increased protein levels and phosphorylation of Thr 308 of hepatic Akt (Fig. 1, lanes D1,2,3).

Fig. 1

Phosphorylation of Akt in the liver following administration of insulin and GC. Four groups of leptin-deficient ob/ob mice were orally administered for four weeks: vehicle (group A), 2.5 mg/kg GC (group B), 2.5 IU of regular insulin (Humulin R, Lilly, group C), and the combination of both GC and insulin (group D). Phosphorylation of Akt in the liver was determined and compared with the actin control. A synergistic effect for the combination therapy was observed (Lanes D1-D3). Proteins were detected with phospho-specific Akt (Cell Signaling, USA, #9271); β-actin (Abcam, UK, sc-47778)

/f/fulltexts/CEH/50787/CEH-9-50787-g001_min.jpg

Effects of oral insulin and GC on serum levels of TNF-a

Serum TNF-α levels varied from 242 pg/ml in group A to 264, 200, and 215 pg/ml in groups B, C, and D, respectively (p = NS, for B vs. A; and p < 0.05, for C and D vs. A) (Fig. 2).

Fig. 2

Serum TNF-α levels following administration of insulin and GC. ELISA measured serum TNF-α levels in all mice in all groups. Oral administration of insulin was associated with a decrease in TNF-α serum levels (p = NS, for B vs. A; and p < 0.05, for C and D vs. A)

/f/fulltexts/CEH/50787/CEH-9-50787-g002_min.jpg

Oral insulin and GC alleviated glucose intolerance and hyperlipidemia

Oral administration of insulin and GC, particularly in combination, significantly reduced the areas under the curve (AUC) of the GTT. AUC was 45649 in group A vs. 25759, 24916, and 19950 in groups B, C, and D, respectively (p ≤ 0.05 for B, C, and D vs. A) (Fig. 3A). GTT was reduced in mice that received the combination of GC and insulin compared to mice that received either GC or insulin (p < 0.01, for D vs. B and C).

Fig. 3

Effects of oral administration of insulin and GC on glucose tolerance and hyperlipidemia. Mice in all experimental groups underwent a glucose tolerance test (GGT), and data are shown for areas under the curve. Following both oral insulin and oral GC, GGT decreased (A). A synergistic effect of the combination therapy with insulin and GC was observed (p ≤ 0.05 for B, C, and D vs. A; p < 0.01 for D vs. B and C). Both oral insulin and oral GC were associated with decreases in serum triglyceride (TG) levels (B, p < 0.05, for B vs. A) and with reductions in serum total cholesterol (C, p < 0.05, for B vs. A; and p < 0.005, for C and D vs. A). The changes were more significant in insulin-treated mice

/f/fulltexts/CEH/50787/CEH-9-50787-g003_min.jpg

Serum TG was significantly lower in all treated groups than in vehicle-treated mice (Fig. 3B, 129, 125, and 116 mg/dl in groups B, C, and D, respectively, compared to 160 mg/dl in group A, p < 0.00005).

Four weeks after initiation of the trial, total serum cholesterol levels were 341 mg/dl in group A vs. 254, 207, and 183 mg/dl in groups B, C, and D, respectively;differences were more profound in insulin-treated mice (Fig. 3C, p < 0.05, for B vs. A; and p < 0.005, for C and D vs. A).

Effects of oral insulin and GC on ALT levels

As early as week 2 of the study, ALT levels were lower in groups B, C, and D (p < 0.01) than in group A. After four weeks of treatment, more significant differences were observed between the groups (420 in group A vs. 215,188, and 168 u/l in groups B, C, and D, respectively; differences were more significant in insulin-treated mice (p < 0.005, for B vs. A; and p < 0.001, for C and D vs. A). The hepatic TG content was reduced by oral insulin and by the combination of insulin and GC, but not by GC alone, 10.4% in group A vs. 10.5%, 5.8%, and 7.7% TG in groups B, C, and D, respectively (Fig. 4B, p = NS, for B vs. A; and p < 0.05, for C and D vs. A).

Fig. 4

A) Changes in ALT serum levels following administration of insulin and GC. ALT serum levels were measured in all experimental groups. Decreases in insulin-treated and GC-treated mice were observed as early as the second week of therapy. At the end of four weeks of treatment, further reductions were observed, which were more significant in insulin-treated mice (p < 0.005, for B vs. A; and p < 0.001, for C and D vs. A). The hepatic triglyceride (TG) content was measured in all mice at the end of the four-week treatment period (B). A significant reduction in liver TGs was noted for insulin-treated mice (p = NS, for B vs. A; and p < 0.05, for C and D vs. A)

/f/fulltexts/CEH/50787/CEH-9-50787-g004_min.jpg

Discussion

Oral administration of insulin or GC, and the combination of both, increased Akt expression in the liver and were associated with a trend for alleviating glucose intolerance and ameliorating liver damage in an ob/ob leptin-deficient mouse model of the metabolic syndrome and NAFLD.

The present study results further support the importance of the Akt pathway in alleviating glucose intolerance and the associated hyperlipidemia and liver damage. PI3K/Akt signaling is relevant in skeletal muscle, adipose tissue, the liver, the brain, and the pancreas, suggesting that it can serve as a therapeutic target for obesity and type 2 diabetes mellitus [5].

Alterations in the PI3K/Akt pathway, found in damaged tissues, are linked to obesity, insulin resistance, and type 2 diabetes. Insulin resistance can further impair the PI3K/Akt pathway, thus forming a vicious circle [5]. Anti-diabetic compounds that target hepatic insulin resistance in type 2 diabetes are associated with activated hepatic AMP-activated protein kinase and PI3K/Akt pathways [34]. The anti-diabetic effects of other compounds were associated with the activation of Akt signaling [35].

The combination therapy of GC and insulin showed a synergistic effect for promoting Akt expression in the liver and alleviating glucose intolerance. An adjuvant effect of GC was previously described in the ob/ob model when co-administered with anti-CD3 [31] and with other drugs and vaccines [25, 36]. These effects were attributed to the potential immunomodulatory effect of GSLs [19, 37]. They also support a potential oral-immunotherapy-based mechanism of action recently described for several compounds [21, 25, 29, 37-48].

The use of GSLs as potential activators or inhibitors of Akt may be related to their structure. Mimics of phosphatidyl were synthesized as Akt inhibitors based on a beta-glucoside scaffold. These compounds possessed one or two lipophilic moieties of different lengths at the anomeric position of glucose and an acidic or basic group at C-6 [49].

Similar to previously published data of studies in both animals and humans [19-23, 25-29, 33, 50-59], in the present study, GC exerted a beneficial effect on liver enzymes and metabolic parameters. However, the effect on liver steatosis observed herein was not demonstrated. This discrepancy may be due, at least in part, to differences in environments [50]. Different effects on liver enzymes and steatosis per se may result from different underlying mechanisms of the pathogenesis involved. The findings also corroborate the notion that most patients with steatosis do not develop non-alcoholic steatohepatitis (NASH) and the observation that NASH also occurs in lean patients [60, 61]. Whether the beta glucosylceramide used in the study has any unique structural feature that might make it an activator of Akt is yet to be determined [17, 19-23, 25-27, 52, 62-68].

Orally delivered insulin mimics the natural route of endogenous insulin secreted by the pancreas into the portal vein and directly to the liver [69]. Insulin therapy is central to diabetes management; however, no commercially available oral formulation is available [70].While attempts are being carried out to achieve clinically meaningful results with oral formulations, the present results support the concept that oral insulin exerts a beneficial effect on liver steatosis and liver damage in NAFLD. The data support potent adjuvants that may synergize their effects by working via similar mechanisms, such as promoting Akt phosphorylation and synergistic pathways.

Treatment with insulin was associated with an anti-inflammatory shift suggesting an orally-immunomodulatory mechanism [37, 46]. The beneficial effects of insulin were associated with a reduction in TNF-α serum levels. TNF-α contributes to the pathogenesis of glucose intolerance associated with obesity [41, 71]. HFD induces glucose intolerance and is associated with increased plasma levels of TNF-α and reduced Akt signaling [72]. The alleviation of insulin resistance in adipocytes was associated with glucose uptake, and increased insulin sensitivity in TNF-α-treated adipocytes was associated with expression of the miR-721-PPAR-gamma-PI3K/AKT-GLUT4 signaling pathway [73]. Inhibition of Akt phosphorylation demonstrated effectiveness in ameliorating glucose intolerance caused by TNF-α [71]. TNF-α induced glucose intolerance via inhibition of Akt/eNOS/NO signaling and was suggested as a target for alleviating glucose intolerance and vascular complications associated with obesity-related conditions [72]. The data suggest that further studies are required to delineate the correlation between TNF-α and Akt pathways.

The study did not include an insulin tolerance test (ITT), which can shed further light on the potential effect of the treatment on glucose intolerance. Future studies will include broadened cytokines, cellular, and signaling molecule profiles, as well as pathological studies, which are expected to dissect better the mechanisms underlying the findings. Comparing the effect of basal versus insulin-stimulated states on Akt expression is expected to further explain its role in the pathogenesis of glucose intolerance. Densitometry studies can improve the accuracy of the reported findings and are subject to future studies.

In summary, the present study results establish Akt expression in the liver as a therapeutic target for treating glucose intolerance, hyperlipidemia, and associated liver damage. Oral administration of insulin with GSLs significantly promoted phosphorylation of Akt, alleviating glucose intolerance and NAFLD in an ob/ob leptin-deficient mouse model of the metabolic syndrome and NAFLD. As preliminary data with oral delivery of insulin [74] and oral GC [29] showed promising results in type 2 diabetes and NASH, their combination may provide a potential therapy to be evaluated in further clinical trials.

Disclosure

Y. Ilan is a consultant for Teva; ENZO; Protalix; Betalin Therapeutics; Immuron; SciM; Natural Shield; Oberon Sciences; Tiziana Pharma; Plantylight; Exalenz Bioscience.

The authors declare no conflict of interest.

References

1 

Kitamura T, Ogawa W, Sakaue H, et al. Requirement for activation of the serine-threonine kinase Akt (protein kinase B) in insulin stimulation of protein synthesis but not of glucose transport. Mol Cell Biol 1998; 18: 3708-3717.

2 

Manna P, Jain SK. Hydrogen sulfide and L-cysteine increase phosphatidylinositol 3,4,5-trisphosphate (PIP3) and glucose utilization by inhibiting phosphatase and tensin homolog (PTEN) protein and activating phosphoinositide 3-kinase (PI3K)/serine/threonine protein kinase (AKT)/protein kinase Czeta/lambda (PKCzeta/lambda) in 3T3l1 adipocytes. J Biol Chem 2011; 286: 39848-39859.

3 

Fuentes M, Santander N, Cortes V. Insulin increases cholesterol uptake, lipid droplet content, and apolipoprotein B secretion in CaCo-2 cells by upregulating SR-BI via a PI3K, AKT, and mTOR-dependent pathway. J Cell Biochem 2019; 120: 1550-1559.

4 

Song C, Liu D, Yang S, et al. Sericin enhances the insulin-PI3K/AKT signaling pathway in the liver of a type 2 diabetes rat model.Exp Ther Med 2018; 16: 3345-3352.

5 

Huang X, Liu G, Guo J, et al. The PI3K/AKT pathway in obesity and type 2 diabetes. Int J Biol Sci 2018; 14: 1483-1496.

6 

Ren L, Zhou X, Huang X, et al. The IRS/PI3K/Akt signaling pathway mediates olanzapine-induced hepatic insulin resistance in male rats. Life Sci 2019; 217: 229-236.

7 

Whelan SA, Dias WB, Thiruneelakantapillai L, et al. Regulation of insulin receptor substrate 1 (IRS-1)/AKT kinase-mediated insulin signaling by O-Linked beta-N-acetylglucosamine in 3T3-L1 adipocytes. J Biol Chem 2010; 285: 5204-5211.

8 

Choi K, Kim YB. Molecular mechanism of insulin resistance in obesity and type 2 diabetes. Korean J Intern Med 2010; 25: 119-129.

9 

Tao A, Xu X, Kvietys P, et al. Experimental diabetes mellitus exacerbates ischemia/reperfusion-induced myocardial injury by promoting mitochondrial fission: Role of down-regulation of myocardial Sirt1 and subsequent Akt/Drp1 interaction. Int J Biochem Cell Biol 2018; 105: 94-103.

10 

Liu C, QiNan W, XiaoTian L, et al. TERT and Akt are involved in the Par-4-dependent apoptosis of islet beta cells in type 2 diabetes. J Diabetes Res 2018; 2018: 7653904.

11 

Pan Y, Wang B, Zheng J, et al. Pancreatic fibroblast growth factor 21 protects against type 2 diabetes in mice by promoting insulin expression and secretion in a PI3K/Akt signaling-dependent manner. J Cell Mol Med 2019; 23: 1059-1071.

12 

Contreras JL, Smyth CA, Bilbao G, et al. Simvastatin induces activation of the serine-threonine protein kinase AKT and increases survival of isolated human pancreatic islets. Transplantation 2002; 74: 1063-1069.

13 

Kubota H, Uda S, Matsuzaki F, et al. In vivo decoding mechanisms of the temporal patterns of blood insulin by the insulin-AKT pathway in the liver. Cell Syst 2018; 7: 118-128 e3.

14 

Wang CY, Kim HH, Hiroi Y, et al. Obesity increases vascular senescence and susceptibility to ischemic injury through chronic activation of Akt and mTOR. Sci Signal 2009; 2: ra11.

15 

Zhou W, Rowitz BM, Dailey MJ. Insulin/IGF-1 enhances intestinal epithelial crypt proliferation through PI3K/Akt, and not ERK signaling in obese humans. Exp Biol Med (Maywood) 2018; 243: 911-916.

16 

Yu T, Gao M, Yang P, et al. Insulin promotes macrophage phenotype transition through PI3K/Akt and PPAR-gamma signaling during diabetic wound healing. J Cell Physiol 2019; 234: 4217-4231.

17 

Ilan Y. Compounds of the sphingomyelin-ceramide-glycosphingolipid pathways as secondary messenger molecules: new targets for novel therapies for fatty liver disease and insulin resistance. Am J Physiol Gastrointest Liver Physiol 2016; 310: G1102-1117.

18 

Fedoryszak-Kuska N, Panasiewicz M, Domek H, et al. Glucosylceramide synthase inhibitors D-PDMP and D-EtDO-P4 decrease the GM3 ganglioside level, differ in their effects on insulin receptor autophosphorylation but increase Akt1 kinase phosphorylation in human hepatoma HepG2 cells. Acta Biochim Pol 2016; 63: 247-251.

19 

Ilan Y. Beta-glycosphingolipids as mediators of both inflammation and immune tolerance: A manifestation of randomness in biological systems. Front Immunol 2019; 10: 1143.

20 

Zigmond E, Zangen SW, Pappo O, et al. Beta-glycosphingolipids improve glucose intolerance and hepatic steatosis of the Cohen diabetic rat. Am J Physiol Endocrinol Metab 2009; 296: E72-78.

21 

Zigmond E, Tayer-Shifman O, Lalazar G, et al. Beta-glycosphingolipids ameliorated non-alcoholic steatohepatitis in the Psammomys obesus model. J Inflamm Res 2014; 7: 151-158.

22 

Zigmond E, Shalev Z, Pappo O, et al. NKT lymphocyte polarization determined by microenvironment signaling: a role for CD8+ lymphocytes and beta-glycosphingolipids. J Autoimmun 2008; 31: 188-195.

23 

Shuvy M, Ben Ya’acov A, Zolotarov L, et al. Beta-glycosphingolipids suppress rank expression and inhibit natural killer T cell and CD8+ accumulation in alleviating aortic valve calcification. Int J Immunopathol Pharmacol 2009; 22: 911-918.

24 

Shu Y, Sheardown SA, Brown C, et al. Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J Clin Invest 2007; 117: 1422-1431.

25 

Mizrahi M, Adar T, Lalazar G, et al. Glycosphingolipids prevent APAP and HMG-CoA reductase inhibitors-mediated liver damage: A novel method for “safer drug” formulation that prevents drug-induced liver injury. J Clin Transl Hepatol 2018; 6: 127-134.

26 

Lalazar G, Ben Ya’acov A, Eliakim-Raz N, et al. Beta-glycosphingolipids-mediated lipid raft alteration is associated with redistribution of NKT cells and increased intrahepatic CD8+ T lymphocyte trapping. J Lipid Res 2008; 49: 1884-1893.

27 

Adar T, Ilan Y. Beta-glycosphingolipids as immune modulators. J Immunotoxicol 2008; 5: 209-220.

28 

Margalit M, Shalev Z, Pappo O, et al. Glucocerebroside ameliorates the metabolic syndrome in OB/OB mice. J Pharmacol Exp Ther 2006; 319: 105-110.

29 

Lalazar G, Zigmond E, Weksler-Zangen S, et al. Oral administration of beta-glucosylceramide for the treatment of insulin resistance and non-alcoholic steatohepatitis: Results of a double-blind, placebo-controlled trial. J Med Food 2017; 20: 458-464.

30 

Paracchini V, Pedotti P, Taioli E. Genetics of leptin and obesity: A HuGE Review. Am J Epidemiol 2005; 162: 101-114.

31 

Ilan Y, Maron R, Tukpah AM, et al. Induction of regulatory T cells decreases adipose inflammation and alleviates insulin resistance in ob/ob mice. Proc Natl Acad Sci U S A 2010; 107: 9765-9770.

32 

Folch J, Lees M, Sloane Stanley GH. A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem 1957; 226: 497-509.

33 

Ben Ya’acov A, Lalazar G, Livovsky DM, et al. Decreased STAT-1phosphorylation by a thio analogue of beta-D-glucosylceramide is associated with altered NKT lymphocyte polarization. Mol Immunol 2009; 47: 526-533.

34 

Yan J, Wang C, Jin Y, et al. Catalpol ameliorates hepatic insulin resistance in type 2 diabetes through acting on AMPK/NOX4/PI3K/AKT pathway. Pharmacol Res 2018; 130: 466-480.

35 

Sun W, Yang J, Wang W, et al. The beneficial effects of Zn on Akt-mediated insulin and cell survival signaling pathways in diabetes. J Trace Elem Med Biol 2018; 46: 117-127.

36 

Shouval D, Ilan Y, Adler R, et al. Improved immunogenicity in mice of a mammalian cell-derived recombinant hepatitis B vaccine containing pre-S1 and pre-S2 antigens as compared with conventional yeast-derived vaccines. Vaccine 1994; 12: 1453-1459.

37 

Ilan Y. Immune rebalancing by oral immunotherapy: A novel method for getting the immune system back on track. J Leukoc Biol 2019; 105: 463-472.

38 

Ben Ya’acov A, Lichtenstein Y, Zolotarov L, et al. The gut microbiome as a target for regulatory T cell-based immunotherapy: induction of regulatory lymphocytes by oral administration of anti-LPS enriched colostrum alleviates immune mediated colitis. BMC Gastroenterol 2015; 15: 154.

39 

Khoury T, Ben Ya’acov A, Shabat Y, et al. Altered distribution of regulatory lymphocytes by oral administration of soy-extracts exerts a hepatoprotective effect alleviating immune mediated liver injury, non-alcoholic steatohepatitis and insulin resistance. World J Gastroenterol 2015; 21: 7443-7456.

40 

Ilan Y. Oral immune therapy: targeting the systemic immune system via the gut immune system for the treatment of inflammatory bowel disease. Clin Transl Immunology 2016; 5: e60.

41 

Ilan Y, Ben Ya’acov A, Shabbat Y, et al. Oral administration of a non-absorbable plant cell-expressed recombinant anti-TNF fusion protein induces immunomodulatory effects and alleviates non-alcoholic steatohepatitis. World J Gastroenterol 2016; 22: 8760-8769.

42 

Almon E, Khoury T, Drori A, et al. An oral administration of a recombinant anti-TNF fusion protein is biologically active in the gut promoting regulatory T cells: Results of a phase I clinical trial using a novel oral anti-TNF alpha-based therapy. J Immunol Methods 2017; 446: 21-29.

43 

Drori A, Rotnemer-Golinkin D, Avni S, et al. Attenuating the rate of total body fat accumulation and alleviating liver damage by oral administration of vitamin D-enriched edible mushrooms in a diet-induced obesity murine model is mediated by an anti-inflammatory paradigm shift. BMC Gastroenterol 2017; 17: 130.

44 

Drori A, Rotnemer-Golinkin D, Zolotarov L, et al. Oral administration of CardioAid and Lunasin alleviates liver damage in a high-fat diet non-alcoholic steatohepatitis model. Digestion 2017; 96: 110-118.

45 

Khoury T, Rotnemer-Golinkin D, Shabat Y, et al. Oral co-administration of soy-derived extracts with alcohol or with sugarsweetened beverages exerts liver and sugar protective effects. J Clin Transl Hepatol 2017; 5: 208-215.

46 

Ilan Y, Shailubhai K, Sanyal A. Immunotherapy with oral administration of humanized anti-CD3 monoclonal antibody: a novel gut-immune system-based therapy for metaflammation and NASH. Clin Exp Immunol 2018; 193: 275-283.

47 

Mizrahi M, Ben Ya’acov A, Adar T, et al. Oral administration of hoodia parviflora alleviates insulin resistance and non-alcoholic steatohepatitis. J Med Food 2019; 22: 1189-1198.

48 

Ishay Y, Potruch A, Weksler-Zangen S, et al. Augmented antiviral T cell immunity by oral administration of IMM-124E in preclinical models and a phase I/IIa clinical trial: A method for the prevention and treatment of COVID-19. Drug Dev Res 2022; 83: 615-621.

49 

Costa B, Dangate M, Vetro M, et al. Synthetic sulfoglycolipids targeting the serine-threonine protein kinase Akt. Bioorg Med Chem 2016; 24: 3396-3405.

50 

Livovsky DM, Lalazar G, Ben Ya’acov A, et al. Administration of beta-glycolipids overcomes an unfavorable nutritional dependent host milieu: a role for a soy-free diet and natural ligands in intrahepatic CD8+ lymphocyte trapping and NKT cell redistribution. Int Immunopharmacol 2008; 8: 1298-1305.

51 

Lalazar G, Ben Ya’acov A, Lador A, et al. Modulation of intracellular machinery by beta-glycolipids is associated with alteration of NKT lipid rafts and amelioration of concanavalin-induced hepatitis. Mol Immunol 2008; 45: 3517-3525.

52 

Lalazar G, Preston S, Zigmond E, et al. Glycolipids as immune modulatory tools. Mini Rev Med Chem 2006; 6: 1249-1253.

53 

Khoury T, Ishay Y, Rotnemer-Golinkin D, et al. A synergistic effect of Ambroxol and beta-glucosylceramide in alleviating immune-mediated hepatitis: A novel immunomodulatory non-immunosuppressive formulation for treatment of immune-mediated disorders. Biomed Pharmacother 2020; 132: 110890.

54 

Shabat Y, Ilan Y. A synergistic effect of Cremophor and beta glucosylceramide to exert liver and sugar protection. J Food Sci Technol 2017; 54: 1184-1191.

55 

Ishay Y, Zimran A, Szer J, et al. Combined beta-glucosylceramide and ambroxol hydrochloride in patients with Gaucher related Parkinson disease: From clinical observations to drug development. Blood Cells Mol Dis 2018; 68: 117-120.

56 

Ben Ya’acov A, Lalazar G, Zolotaryova L, et al. Impaired liver regeneration by beta-glucosylceramide is associated with decreased fat accumulation. J Dig Dis 2013; 14: 425-432.

57 

Zhang W, Moritoki Y, Tsuneyama K, et al. Beta-glucosylceramide ameliorates liver inflammation in murine autoimmune cholangitis. Clin Exp Immunol 2009; 157: 359-364.

58 

Safadi R, Zigmond E, Pappo O, et al. Amelioration of hepatic fibrosis via beta-glucosylceramide-mediated immune modulation is associated with altered CD8 and NKT lymphocyte distribution. Int Immunol 2007; 19: 1021-1029.

59 

Zigmond E, Preston S, Pappo O, et al. Beta-glucosylceramide: a novel method for enhancement of natural killer T lymphoycte plasticity in murine models of immune-mediated disorders. Gut 2007; 56: 82-89.

60 

Araujo AR, Rosso N, Bedogni G, et al. Global epidemiology of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: What we need in the future. Liver Int 2018; 38 Suppl 1: 47-51.

61 

Das K, Chowdhury A. Lean NASH: distinctiveness and clinical implication. Hepatol Int 2013; 7 Suppl 2: 806-813.

62 

Adar T, Ben Ya’acov A, Shabat Y, et al. Steroid-mediated liver steatosis is CD1d-dependent, while steroid-induced liver necrosis, inflammation, and metabolic changes are CD1d-independent. BMC Gastroenterol 2022; 22: 169.

63 

Adar T, Shankar Lankalapalli R, Bittman R, et al. The assembly of glycosphingolipid determines their immunomodulatory effect: A novel method for structure-based design of immunotherapy. Cell Immunol 2020; 355: 104157.

64 

Ilan-Ber T, Ilan Y. The role of microtubules in the immune system and as potential targets for gut-based immunotherapy. Mol Immunol 2019; 111: 73-82.

65 

Ilan Y. Alpha versus beta: are we on the way to resolve the mystery as to which is the endogenous ligand for natural killer T cells? Clin Exp Immunol 2009; 158: 300-307.

66 

Lalazar G, Ben Ya’acov A, Livovsky DM, et al. Beta-glycoglycosphingolipid-induced alterations of the STAT signaling pathways are dependent on CD1d and the lipid raft protein flotillin-2. Am J Pathol 2009; 174: 1390-1399.

67 

Mizrahi M, Lalazar G, Ben Ya’acov A, et al. Beta-glycoglycosphingolipid-induced augmentation of the anti-HBV immune response is associated with altered CD8 and NKT lymphocyte distribution: a novel adjuvant for HBV vaccination. Vaccine 2008; 26: 2589-2595.

68 

El Haj M, Ben Ya’acov A, Lalazar G, et al. Potential role of NKT regulatory cell ligands for the treatment of immune mediated colitis. World J Gastroenterol 2007; 13: 5799-5804.

69 

Eldor R, Kidron M, Arbit E. Open-label study to assess the safety and pharmacodynamics of five oral insulin formulations in healthy subjects. Diabetes Obes Metab 2010; 12: 219-223.

70 

Lopes M, Simoes S, Veiga F, et al. Why most oral insulin formulations do not reach clinical trials. Ther Deliv 2015; 6: 973-987.

71 

Ikubo M, Wada T, Fukui K, et al. Impact of lipid phosphatases SHIP2 and PTEN on the time-and Akt-isoform-specific amelioration of TNF-alpha-induced insulin resistance in 3T3-L1 adipocytes. Am J Physiol Endocrinol Metab 2009; 296: E157-164.

72 

da Costa RM, Neves KB, Mestriner FL, et al. TNF-alpha induces vascular insulin resistance via positive modulation of PTEN and decreased Akt/eNOS/NO signaling in high fat diet-fed mice. Cardiovasc Diabetol 2016; 15: 119.

73 

Ke B, Ke X, Wan X, et al. Astragalus polysaccharides attenuates TNF-alpha-induced insulin resistance via suppression of miR-721 and activation of PPAR-gamma and PI3K/AKT in 3T3-L1 adipocytes. Am J Transl Res 2017; 9: 2195-2206.

74 

Wong CY, Al-Salami H, Dass CR. Potential of insulin nanoparticle formulations for oral delivery and diabetes treatment. J Control Release 2017; 264: 247-275.

Copyright: © 2023 Clinical and Experimental Hepatology. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.