research article

Exenatide Prevents Diet-induced Hepatocellular Injury in A CEACAM1-Dependent Mechanism

Hilda E. Ghadieh1, Harrison T.Muturi2, Sonia M.Najjar1,2,3*

1Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA

2Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA

3Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA      

*Corresponding author: Sonia M. Najjar, Department of Biomedical Sciences; Irvine Hall, Ohio University, Athens, OH 45701-2979, USA. Tel: +17405932376; Fax:+17405932778; Email: najjar@ohio.edu

Received Date: 27 November, 2017; Accepted Date: 11 December, 2017; Published Date: 19 December, 2017

 Citation: Ghadieh HE, Muturi HT, Najjar SM (2017) Exenatide Prevents Diet-induced Hepatocellular Injury in A CEACAM1-Dependent Mechanism. J Diabetes Treat: 133. DOI: 10.29011/2574-7568.000033

1.      Abstract

The Carcinoembryonic Antigen-Related Cell Adhesion Molecule1 (CEACAM1) promotes insulin sensitivity by inducing insulin clearance and reducing de novo lipogenesis in the liver. Consistently, Cc1–/–mice with null deletion of Ceacam1 gene exhibit hyperinsulinemia and insulin resistance, in addition to steatohepatitis. They also exhibit early pericellular fibrosis. Redelivering Ceacam1 to the liver reverses the altered metabolism and histopathology of Cc1–/– mice. Exenatide, a long-acting glucagon-like peptide-1 receptor agonist, induces Ceacam1 transcription and consequently, reverses impaired insulin clearance and insulin resistance caused by high-fat intake.Additionally, it reverses fat accumulation in the liver. The current studies show that exenatide also restored the activities of alanine transaminase and aspartate aminotransferase, and reversed the inflammatory and oxidative stress response to high-fat diet in wild-type, but not in Cc1–/– mice. Exenatide also prevented diet-induced activation of the TGFb/Smad2/Smad3 pro-fibrogenic pathways, and normalized the mRNA levels of pro-fibrogenic genes in wild-type, but not in Cc1–/– mice. Together, the data demonstrate that exenatide prevented diet-induced pro-fibrogenesis and hepatocellular injury in a CEACAM1-dependent mechanism. 

2.      Keywords: Fibrosis; Glucagon-Like Peptide-1; Insulin Clearance; Insulin Resistance; Steatohepatitis

1.      Nonstandard Abbreviations

ALT                        :               Alanine Transaminase

AST                        :               Aspartate Aminotransferase

CEACAM1           :               Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1

Cc1–/–                     :               Global Ceacam1 Null Mouse

Cc1+/+                     :               Wild-Type Littermate of Cc1–/–

GLP-1                    :               Glucagon-Like Peptide-1

RD                          :               Regular Diet

HF                          :               High-Fat Diet

2.      Introduction

Nonalcoholic Fatty Liver Disease (NAFLD) ranges from benign steatosis to steatohepatitis and Non-Alcoholic Steatohepatitis (NASH) that also includes chicken-wire bridging fibrosis [1].Uncontrolled, the disease can progress to adenocarcinoma to constitute a major risk factor for liver transplant [2].

The paucity of mouse models that replicate faithfully NASH human disease and in particular fibrosis [3], has limited our understanding of its molecular underpinning and consequently, has restricted progress in the development of effective pharmacologic interventions. Studies in our laboratory have identified a role for the Carcinoembryonic Antigen-Related Cell Adhesion molecule 1 (CEACAM1) in inducing hepatic insulin clearance to promote insulin sensitivity and reduce hepatic de novo lipogenesis to protect the liver against the high level of insulin in the portal circulation [4]. Accordingly, mice with global null deletion of Ceacam1 gene (Cc1–/–)[5] and with liver-specific inactivation of CEACAM1 [6] develop impaired insulin clearance, followed by hyperinsulinemia and insulin resistance, in addition to fat accumulation in the liver, largely resulting from hyperinsulinemia-induced activation of the transcription of lipogenic genes [7,8]. With increased fat storage triggering changes in the inflammatory milieu [9,10], Ceacam1 mutants also develop steatohepatitis.Moreover, they develop a NASH-characteristic chicken-wire pattern of fibrosis on regular chow diet [5]. When fed a high-fat diet, these features of hepatocyte injury progress to include advanced chicken-wire fibrosis and apoptosis [11,12].

Recently, we have found that exenatide, a long-acting Glucagon-Like Peptide-I (GLP-I) receptor agonist and a synthetic analog of Exendin-4 that induces insulin secretion in part by inhibiting glucagon secretion [13,14], also induces CEACAM1-dependent hepatic insulin clearance [15]. Whereas exenatide reverses steatohepatitis in wild-type mice fed a high-fat diet, it fails to do so in Cc1–/– mice. The underlying mechanism involves the induction of Ceacam1 expression by binding directly to the peroxisome Proliferator-Activated Receptor Response Element (PPRE)/retinoid X receptor-a (RXRa) on the Ceacam1 promoter, and activating its transcription [15]. Thus, we aimed in the current study to investigate whether exenatide also ameliorates fibrosis, and whether this requires intact CEACAM1 expression.

3.      Materials and Methods

3.1.  Mice Maintenance

C57BL/6.Cc1−/− and Cc1+/+ littermates (3 months of age) were fed ad libitum a standard (RD) or a High-Fat (HF) diet deriving 45:35:20% calories from fat:carbohydrate:protein (D12451, Research Diets) for 2 months [16]. In the last month of feeding, mice received an intraperitoneal injection/day of saline or exenatide (20ng/g BW/day) (507-77, California Peptide Research, Salt Lake City, UT) [15]. All procedures were approved by the Institutional Animal Care and Utilization Committee at the University of Toledo.

3.2.  ALT and AST Colorimetric Assays

Per manufacturer’s instructions (Abcam, Cambridge, MA), liver tissues (50mg) were homogenized in 200µl assay buffers, centrifuged (13,000xg, 10 min), and aliquots from the supernatant layer were added to 100µl of the reaction mix in the Alanine Transaminase (ALT) (ab105134)and Aspartate Aminotransferase (AST) (ab105135) kits. The products were read at OD570nm, and activities were measured in µM/mgmU/mg..

3.3.  Western Blot Analysis

Livers were lysed and proteins analyzed by SDS-PAGE followed by immunoprobing with polyclonal antibodies against phospho-Smad2Ser465/467 and phospho-Smad3Ser423/425 (Cell signaling, Danvers, MA). For normalization, proteins were reprobed with polyclonal antibodies against Smad2 and Smad3 (Cell signaling). Blots were incubated with horseradish peroxidase-conjugated donkey anti-rabbit IgG antibody (GE Healthcare Life Sciences, Amersham, Marlborough, MA) and proteins were visualized using ECL (Amersham).

3.4.  Liver Histology

As described [12], Formalin-fixed, paraffin-embedded liver sections were stained with 0.1% Sirius Red stain (Sigma, Direct Red 80). Fibrosis was assessed on deparaffinized and rehydrated slides and scored using the Brunt Criteria [17,18].

3.5.  Quantitative Real-time PCR Analysis

PerfectPure RNA Tissue Kit (5 PRIME Inc.) was used to isolated total RNA and cDNA was synthesized by iScript cDNA Synthesis Kit (BIO-RAD), using 1μg of total RNA and oligo-dT primers (Table 1).cDNA was evaluated with qRT-PCR (StepOne Plus, Applied Biosystems) and normalized to 18S. Results are expressed in fold change as the mean±SEM.

3.6.  Statistical Analysis

Data were analyzed by one-way ANOVA or the two-tailed Student-t-test using GraphPad Prism 6 software. P<0.05 was considered statistically significant.

4.      Results

4.1.  Effect of Exenatide on Hepatic ALT and AST Activities

Cc1–/– mice exhibited higher hepatic ALT (Figure 1A) and AST (Figure 1B) activities than their age-matched Cc1+/+wild-type counterparts, as previously reported [19]. HF feeding for 2 months elevated hepatic ALT and AST activities in Cc1+/+ and Cc1–/– mice (Figure. 1A and 1B, HF-S vs RD-S). Treating with exenatide in the last 30 days of HF feeding reversed this increase in Cc1+/+, but not Cc1–/– mice (Figure 1A and 1B, HF-Ex vs HF-S).

4.2.  Effect of Exenatide on Oxidative Stress and Inflammation

qRT-PCR analysis revealed that HF diet induced hepatic mRNA levels of markers of oxidative stress, including Nox1, Nox 4 and Gp91, by ~2- to 3-fold in both groups of mice (Table 2, HF-S vs RD-S), as expected [20]. This was accompanied by a 2-fold increase in mRNA levels of markers of inflammation (Il-6, IFNg and TNFa) (Table 2, HF-S vs RD-S). Exenatide treatment reversed the positive effect of HF diet on the hepatic mRNA levels of the inflammatory and oxidative stress markers in Cc1+/+, but not Cc1–/– mice (Table 2, HF-Ex vs HF-S). That exenatide reversed inflammation in Cc1+/+, but not Cc1–/–mice, is supported by our previously published H&E stain analysis [15].

4.3.  Effect of Exenatide on Hepatic Fibrosis

qRT-PCR analysis showed higher mRNA levels of pro-fibrotic genes (Col6-a3, a-Sma and TGFb) and lower mRNA levels of Smad7, an inhibitor of TGFb activation, in the liver of untreated HF-fed relative to RD-fed Cc1+/+ mice (Table 1, HF-S vs RD-S). Accordingly, Western blot analysis revealed induction of Smad2/Smad3 phosphorylation by HF diet in Cc1+/+mice (Figure2A, HF-S vs RD-S). Sirius red staining showed periportal fibrosis in HF-S Cc1+/+mice (Figure 2Bii and a Brunt score of 2-accompanying table). Exenatide treatment reversed these HF-induced profibrogenic parameters in Cc1+/+ mice (Table 2 and Figure2A, HF-Ex vs S-Ex, and Figure2Biv with a Brunt score of 0). As expected from our previous studies [12], Sirius red staining revealed perivenular and/or pericellular bridging chicken-wire pattern of collagen deposition in the liver of RD- and HF-fed Cc1−/− mice (Figure2Bv-vi and a Brunt score of 3). This NASH-like fibrosis in Cc1−/− mice, whether spontaneously (under RD feeding conditions-Figure2Bv) or in response to HF diet (Figure 2Bvi) was not reversed by 4 weeks of exenatide treatment (Figure2Bvii and Figure2Bviii, respectively and a Brunt score of 3). Consistently, exenatide failed to normalize the mRNA levels of markers of fibrosis modulators under both feeding conditions (Table 2, RD/HF-Ex vs RD/HF-S), as well as it failed to reduce Smad2/Smad3 activation by HF diet in Cc1–/–mice (Figure 2A, RD/HF-Ex vs RD/HF-S).

5.      Discussion

Cc1–/– mice develop insulin resistance, steatohepatitis with spontaneous chicken-wire fibrosis that become more robust in response to high-fat feeding [12]. Liver-specific rescuing of CEACAM1 expression reverses hyperinsulinemia, insulin resistance, steatohepatitis and visceral obesity in Cc1–/– mice [21]. We have also shown that high-fat feeding for 21 days reduces CEACAM1 expression by >50% in C56BL/6 mice to cause insulin resistance and metabolically phenocopy the Cc1–/– mouse [16], but forced liver-specific overexpression [16] and adenoviral-mediated redelivery of CEACAM1 [22] prevents these metabolic abnormalities together with the rise in profibrogenic genes in HF-fed wild-type mice. Together, this suggests that loss of CEACAM1 in liver plays a critical role in the pathogenesis of metabolic and histological abnormalities detected in NAFLD.In support of this proposed CEACAM1-based mechanism, exenatide reverses insulin resistance and steatohepatitis in wild-type, but not Cc1–/– mice, via inducing Ceacam1 transcription [15]. The current studies showed that exenatide also reversed the other features of hepatocyte injury caused by high-fat feeding in Cc1+/+, but not Cc1–/– mice. These include: activation of ALT and AST, oxidative stress as assessed by changes in the mRNA levels of associated genes, and of the TGFb-mediated profibrogenic pathways in liver.

Normalization of ALT and AST activities, in addition to preventing the rise in inflammatory markers and inactivation of TGFb pathways by exenatide is consistent with the ameliorating effect of IP118, a GLP-1 receptor agonist, on these markers of hepatocyte injury in high-fat diet-fed C57BL/6 mice [23]. Failure of exenatide to modulate these metabolic and pathological phenotypes in RD- and HF-fed Cc1–/– mice points to the key role of CEACAM1 in mediating the beneficial effects of exenatide. Thus, it is likely that by binding to the PPRE/RXR element on the Ceacam1 promoter [15], exenatide induces CEACAM1 expression in liver to prevent the advancement of hepatocyte injury, including the increase in fat accumulation and inflammation caused by high-fat diet [24].

In addition to basal steatosis and inflammation, Cc1–/– mice exhibit low levels of pericellular fibrosis when fed a regular chow diet [12]. The current studies show that this is accompanied by basal induction of Smad2/Smad3 in the TGFbpathway, which similarly occurs in the liver of C57BL/6 mice fed a high-fat diet, likely resulting from the >50% decrease in hepatic CEACAM1 level by high-fat intake, as we have previously shown [16]. Reversal of these pathways by adenoviral-mediated liver-specific redelivery of CEACAM1 demonstrates the anti-fibrogenic role of hepatic CEACAM1 in the liver [22] as well as in the white adipose tissue [25]. Inactivation of the TGFb fibrogenic pathway by exenatide in Cc1+/+,but not Cc1–/– mice, further assigns a critical role for hepatic CEACAM1 induction in the anti-fibrogenic effect of exenatide.Mechanistically, the loss of hepatic CEACAM1 impairs insulin clearance to cause hyperinsulinemia, followed by insulin resistance and elevated hepatic de novo lipogenesis [8], both being risk factors for fibrosis [26]. Increased fat accumulation can change the inflammatory microenvironment [9,10] in the liver to release profibrogenic factors [27], and proinflammatory cytokines [28] that modify hepatic inflammation and contribute to fibrosis [24].

Moreover, increase in hepatic lipid production yields redistribution of substrates to the white adipose tissue to cause visceral adiposity followed by associated induction of the pro-inflammatory state, including the release of leptin, which can exacerbate the fibrogenic effect of TNFa[29; 30], which causes oxidative stress [31] and reduces Smad7 expression [32], leading to the activation of the TGFb/Smad2/Smad3 fibrogenic pathways. In addition to inducing the activity of TGFb, high-fat diet also increases its hepatic level together with that of the pro-fibrogenic factor, IL-6 [33,34], but with a rise in the anti-fibrogenic IFNg[34]. With exenatide inducing hepatic CEACAM1 production that contributes substantially to the decrease in visceral obesity [15], and subsequently, the pro-inflammatory state associated with high-fat feeding, it is conceivable that induction of CEACAM1 expression is required for the beneficial effect of exenatide not only in restoring the metabolic phenotype caused by high-fat intake, but also in limiting the progression of fibrosis.

6.      Conclusion

Our data emphasize that induction of hepatic CEACAM1 by exenatide mediates its effect not only on insulin resistance, hepatic steatosis and visceral obesity [15], but also on the production of IL-6 and TNFa, which would, in turn limit their pro-inflammatory and pro-fibrogenic effect. The significance of this finding to human disease is highlighted by the reported low hepatic CEACAM1 levels in insulin resistance obese patients with fatty liver disease [35]. While by inducing CEACAM1 expression and promoting hepatic insulin clearance, exenatide maintains insulin levels at physiologic levels in the face of increased insulin secretion[15] in order to limit insulin resistance and steatohepatitis in animal models of NAFLD/NASH [36-38], the data supporting its clinical relevance and safety in the treatment of NASH remain limited [39] and warrant further investigations.

7.      Acknowledgements

The authors thank Melissa W. Kopfman and Zachary N. Smiley at the Najjar Laboratory at the University of Toledo College of Medicine for their technical assistance in exenatide injections, and in the generation and maintenance of mice. This work was supported by grants from the National Institutes of Health; R01-DK054254, R01-DK083850, and R01-HL112248 to SMN, and by fellowships from the Middle-East Diabetes Research Center to HEG. The studies were also partially supported by the John J. Kopchick PhD Ohio Heritage Foundation Eminent Research Chair Fund to SMN.

o


Figure 1: Effect of exenatide on hepatic ALT and AST activities. Mice were fed with a Regular Diet (RD) or a High-Fat Diet (HF) for 2 months and injected daily with either Saline (S) or Exenatide (Ex) in the last month of feeding. At the end of the feeding/treatment period, AST and ALT activities were measured in duplicate in liver lysates (n=5 mice/ genotype/ feeding/ treatment). RD-S (white), RD-Ex (light grey), HF-S (dark grey), and HF-Ex (black). Values are expressed as mean±SEM. *P<0.05 HF vs RD/ treatment group; P<0.05 Ex vs S/ feeding group; P<0.05 Cc1−/− vs Cc1+/+ mice/ feeding group/ treatment group.




Figure 2: Effect of exenatide on hepatic fibrosis. A. Liver lysates were analyzed by immunoblotting with α-phospho-Smad2 (α-pSmad2) and α-phospho-Smad3 (α-pSmad3) antibodies followed by reimmunoprobing (reIb) with antibodies against total Smad2 and total Smad3, respectively, for normalization. Gels represent more than 2 experiments performed on different mice/ feeding/ treatment group.B:  Liver histology were analyzed by Sirius red staining to detect bridging fibrosis in Cc1–/–mice (n=7-8/ genotype/ feeding/ treatment). Representatives from S-treated RD-fed (i and v), S-treated HF-fed (ii and vi),Ex-treated RD-fed (iii and vii); and Ex-treated HF-fed mice (iv and viii) are shown. The degree of fibrosis was evaluated per Brunt Criteria, and scores are included in the accompanying table.

 

Primer

 

Forward Sequence (5'-3')

Reverse Sequence (5'-3')

Col6-a3

GTCAGCTGAGTCTTGTGCTGT

ACCTAGAGAACGTTACCTCACT

a-Sma

CGTGGCTATTCCTTCGTTAC

TGCCAGGAGACTCCATCC

TGFβ

GTGGAAATCAACGGGATCAG

ACTTCCAACCCAGGTCCTTC

Smad7

GTTGCTGTGAATCTTACGGG

ATCTGGACAGCCTGCA

IFNg

ATG AACGCTACACACTGCATC

CCATCCTTTTGCCAGTTCCTC

IL-6

CTTGGGACTGCCGCTGGTGA

TGCAAGTGCATCATCGTTGT

TNFa

CCACCACGCTCTTCTGTCTAC

AGGGTCTGGGCCATAGAACT

Nox1

TTACACGAGAGAAATTCTTGGG

TCGACACACAGGAATCAGGA

Nox4

TCCAAGCTCATTTCCCACAG

CGGAGTTCCATTACATCAGAGG

Gp91

TATGCTGATCCTGCTGCCAGT

TGTCTTCGAATCCTTGTCGAGC

18S

TTCGAACGTCTGCCCTATCAA

ATGGTAGGCACGGCGACTA

 

Table 1: Primer sequence of mouse genes used in quantitative Real-time PCR analysis.

 

 

 RD-S

 RD-Ex

 HF-S

 HF-Ex

a) Cc1+/+

 

 

 

 

Fibrosis

 

 

 

 

Col6-a3

2.11 ± 0.22

2.33 ± 0.20

6.11 ± 0.30*

2.37 ± 0.25

a-Sma

3.02 ± 0.34

3.15 ± 0.31

6.99 ± 0.29*

3.19 ± 0.23

TGFb

2.36 ± 0.26

2.18 ± 0.21

6.16 ± 0.28*

2.13 ± 0.26

Smad7

1.36 ± 0.04

1.41 ± 0.04

0.38 ± 0.06*

1.47 ± 0.04

Oxidative stress

 

 

 

 

Nox1

1.12 ± 0.07

1.21 ± 0.06

3.26 ± 0.12*

1.28 ± 0.11

Nox4

1.08 ± 0.05

1.11 ± 0.07

3.01 ± 0.10*

1.22 ± 0.12

Gp91

1.11 ± 0.12

1.16 ± 0.08

2.98 ± 0.17*

1.12 ± 0.13

Inflammation

 

 

 

 

Il-6

4.46 ± 0.18

4.21 ± 0.11

8.71 ± 0.21*

4.23 ± 0.19

IFNg

7.22 ± 0.18

6.87 ± 0.25

12.5 ± 0.25*

6.88 ± 0.15

TNFa

3.07 ± 0.21

2.86 ± 0.22

6.86 ± 0.22*

2.73 ± 0.11

bCc1–/–

 

 

 

 

Fibrosis

 

 

 

 

Col6-a3

3.84 ± 0.18

4.07 ± 0.33

10.6 ± 0.41*

10.8 ± 0.41*

a-Sma

5.47 ± 0.30

5.68 ± 0.35

12.9 ± 0.44*

12.7 ± 0.42*

TGFb

5.22 ± 0.26

5.47 ± 0.33

12.5 ± 0.47*

12.0 ± 0.49*

Smad7

2.88 ± 0.10

3.09 ± 0.07

1.74 ± 0.10*

1.76 ± 0.07*

Oxidative stress

 

 

 

 

Nox1

1.34 ± 0.11

1.45 ± 0.15

3.87 ± 0.18*

3.58 ± 0.17*

Nox4

1.02 ± 0.12

1.13 ± 0.11

3.57 ± 0.22*

3.48 ± 0.20*

Gp91

1.36 ± 0.22

1.43 ± 0.21

3.03 ± 0.28*

3.11 ± 0.24*

Inflammation

 

 

 

 

Il-6

7.13 ± 0.21

7.43 ± 0.14

13.4 ± 0.31*

12.9 ± 0.34*

IFNg

7.22 ± 0.19

7.41 ± 0.18

14.3 ± 0.21*

14.1 ± 0.27*

TNFa

5.51 ± 0.23

5.74 ± 0.22

11.2 ± 0.27*

11.0 ± 0.21*

Male Mice (3-month-old) were fed RD or HF for 2 months. In the last 30 days of feeding, they were injected intraperitoneally once daily with Saline (S) or Exenatide (Ex) (20ng/g BW) (n=5/genotype/feeding/treatment). Hepatic qRT-PCR analysis was carried out in triplicate and normalized to 18S. Values are expressed as mean±SEM. *P<0.05 HF versus RD/each of S or Ex treatment group; P<0.05 Ex versus S/each of RD or HF feeding group


Table 2: Effect of Exenatide on the mRNA levels of genes in the liver of male mice.

  1. Spengler EK, Loomba R (2015) Recommendations for Diagnosis, Referral for Liver Biopsy, and Treatment of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Mayo Clin Proc 90: 1233-1246.
  2. Ascha MS, Hanouneh IA, Lopez R, Tamimi TA, Feldstein AF, et al.(2010) The incidence and risk factors of hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. Hepatology 51: 1972-1978.
  3. S.K. Erickson (2009) Nonalcoholic fatty liver disease. J. Lipid Res 50: S412-S416.
  4. Ward GM, Walters JM, Aitken PM, Best JD, AlfordFP (1990) Effects of prolonged pulsatile hyperinsulinemia in humans. Enhancement of insulin sensitivity. Diabetes 39: 501-507.
  5. DeAngelis AM, Heinrich G, Dai T, Bowman TA, Patel PR, et al. (2008) Carcinoembryonic antigen-related cell adhesion molecule 1: a link between insulin and lipid metabolism. Diabetes 57: 2296-2303.
  6. Poy MN, Yang Y, Rezaei K, Fernstrom MA, Lee AD, et al. (2002) CEACAM1 regulates insulin clearance in liver. Nat Genet 30: 270-276.
  7. Horton JD, Goldstein JL, Brown MS (2002) SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. The Journal of clinical investigation 109: 1125-1131.
  8. Heinrich G, Ghadieh HE, Ghanem SS, Muturi HT, Rezaei K, et al. (2017) Loss of Hepatic CEACAM1: A Unifying Mechanism Linking Insulin Resistance to Obesity and Non-Alcoholic Fatty Liver Disease. Front Endocrinol 8.
  9. Sheth SG, Gordon FD, Chopra S (1997) Nonalcoholic steatohepatitis. Ann Intern Med 126: 137-145.
  10. Bigorgne AE, Bouchet-Delbos L, Naveau S, Dagher I, Prévot S, et al. (2008) Obesity-induced lymphocyte hyperresponsiveness to chemokines: a new mechanism of Fatty liver inflammation in obese mice. Gastroenterology 134: 1459-1469.
  11. Lee SJ, Heinrich G, Fedorova L, Al-Share QY, Ledford KJ (2008) Development of nonalcoholic steatohepatitis in insulin-resistant liver-specific S503A carcinoembryonic antigen-related cell adhesion molecule 1 mutant mice. Gastroenterology 135: 2084-2095.
  12. Ghosh S, Kaw M, Patel PR, Ledford KJ, Bowman TA, et al, (2010) Mice with null mutation of Ceacam I develop nonalcoholic steatohepatitis. Hepat Med Res Evidence: 69-78.
  13. DruckerDJ(2015) Deciphering metabolic messages from the gut drives therapeutic innovation: the 2014 Banting Lecture. Diabetes 64: 317-326.
  14. D'Alessio D (2016) Is GLP-1 a hormone: Whether and When? J. Diabetes Investig. 7: 50-55.
  15. Ghadieh HE, Muturi HT, Russo L, Marino CC, Ghanem SS, S et al, (2017) Exenatide induces carcinoembryonic antigen-related cell adhesion molecule 1 expression to prevent hepatic steatosis. Hepatololy Commun.
  16. Al-Share QY, DeAngelis AM, Lester SG, Bowman TA, Ramakrishnan SK, et al. (2015) Forced Hepatic Overexpression of CEACAM1 Curtails Diet-Induced Insulin Resistance. Diabetes 64: 2780-2790.
  17. Santiago-Rolón A, Purcell D, Rosado K, Toro DH (2015) A Comparison of Brunt's Criteria, the Non-Alcoholic Fatty Liver Disease Activity Score (NAS), and a Proposed NAS Scoring that Includes Fibrosis in Non-Alcoholic Fatty Liver Disease Staging. PR Health Sci J 34: 189-194.
  18. Brown GT, Kleiner DE (2016) Histopathology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Metabolism 65: 1080-1086.
  19. Xu E, Dubois MJ, Leung N, Charbonneau A, Turbide C, et al. (2009) Targeted disruption of carcinoembryonic antigen-related cell adhesion molecule 1 promotes diet-induced hepatic steatosis and insulin resistance. Endocrinology 150: 3503-3512.
  20. Robertson G, Leclercq I, Farrell GC (2001) Nonalcoholic steatosis and steatohepatitis. II. Cytochrome P-450 enzymes and oxidative stress. Am J Physiol Gastrointest Liver Physiol 281: G1135-G1139.
  21. Russo L1 Muturi HT, Ghadieh HE, Ghanem SS, Bowman TA, et al. (2017) Liver-specific reconstitution of CEACAM1 reverses the metabolic abnormalities caused by its global deletion in male mice. Diabetologia 60: 2463-2474.
  22. Russo L, Ghadieh HE, Ghanem SS, Al-Share QY, Smiley ZN,et al. (2016) Role for hepatic CEACAM1 in regulating fatty acid metabolism along the adipocyte-hepatocyte axis. JLipid Res 57: 2163-2175.
  23. Jouihan H, Will S, Guionaud S, Boland ML, Oldham S, et al. (2017) Superior reductions in hepatic steatosis and fibrosis with co-administration of a glucagon-like peptide-1 receptor agonist and obeticholic acid in mice. Mol Metab 6: 1360-1370.
  24. Najjar SM, Russo L (2014) CEACAM1 loss links inflammation to insulin resistance in obesity and non-alcoholic steatohepatitis (NASH). SeminImmunopathol 36: 55-71.
  25. Lester SG, Russo L, Ghanem SS, Khuder SS, DeAngelis AM, et al. (2015) Hepatic CEACAM1 Over-Expression Protects Against Diet-Induced Fibrosis and Inflammation in White Adipose Tissue. Front Endocrinol (Lausanne) 6: 116-122.
  26. Angulo P, Keach JC, Batts KP, Lindor KD (1999) Independent predictors of liver fibrosis in patients with nonalcoholic steatohepatitis. Hepatology 30: 1356-1362.
  27. Wobser H, Dorn C, Weiss TS, Amann T, Bollheimer C, et al. (2009) Lipid accumulation in hepatocytes induces fibrogenic activation of hepatic stellate cells. Cell Res 19: 996-1005.
  28. Li BH, He FP, Yang X, Chen YW, Fan JG (2017) Steatosis induced CCL5 contributes to early-stage liver fibrosis in nonalcoholic fatty liver disease progress. Transl Res 180: 103-117.
  29. Manco M, Marcellini M, Giannone G, Nobili V (2007) Correlation of Serum TNF-alpha Levels and Histologic Liver Injury Scores in Pediatric Nonalcoholic Fatty Liver Disease. Am J Clin Pathol 127: 954-960.
  30. Carter-Kent C, Zein NN, Feldstein AE (2008) Cytokines in the pathogenesis of fatty liver and disease progression to steatohepatitis: implications for treatment. Am J Gastroenterol 103: 1036-1042.
  31. Crespo J, Cayón A, Fernández-Gil P, Hernández-Guerra M, Mayorga M, et al. (2001) Gene expression of tumor necrosis factor alpha and TNF-receptors, p55 and p75, in nonalcoholic steatohepatitis patients. Hepatology 34: 1158-1163.
  32. Nagarajan RP, Chen F, Li W, Vig E, Harrington MA, et al. (2000) Repression of transforming-growth-factor-beta-mediated transcription by nuclear factor kappaB. Biochem J  348: 591-596.
  33. Syn WK1, Choi SS, Diehl AM (2009) Diehl, Apoptosis and cytokines in non-alcoholic steatohepatitis. Clin Liver Dis 13: 565-580.
  34. Bhogal RK, Bona CA (2005) B cells: no longer bystanders in liver fibrosis. J Clin Invest 115: 2962-2965.
  35. LeeW (2011)The CEACAM1 expression is decreased in the liver of severely obese patients with or without diabetes. Diagn. Pathol 6: 40.
  36. Svegliati-Baroni G, Saccomanno S, Rychlicki C, Agostinelli L, De Minicis S, et al. (2011) Glucagon-like peptide-1 receptor activation stimulates hepatic lipid oxidation and restores hepatic signalling alteration induced by a high-fat diet in nonalcoholic steatohepatitis. Liver Int 31: 1285-1297.
  37. Trevaskis JL, Griffin PS, Wittmer C, Neuschwander-Tetri BA, Brunt EM, et al. (2012) Glucagon-like peptide-1 receptor agonism improves metabolic, biochemical, and histopathological indices of nonalcoholic steatohepatitis in mice. Am JPhysiol Gastrointest Liver Physiol 302: G762-G772.
  38. Ding X1, Saxena NK, Lin S, Gupta NA, Anania FA (2006) Exendin-4, a glucagon-like protein-1 (GLP-1) receptor agonist, reverses hepatic steatosis in ob/ob mice. Hepatology 43: 173-181.
  39. Le TA, Loomba R (2012) Management of Non-alcoholic Fatty Liver Disease and Steatohepatitis. J Clin Exp Hepatol 2: 156-173.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Diabetes and Treatment

cara menggunakan pola slot mahjongrtp tertinggi hari inislot mahjong ways 1pola gacor olympus hari inipola gacor starlight princessslot mahjong ways 2strategi olympustrik mahjong ways 2trik olympus hari inirtp koi gatertp pragmatic tertinggicheat jackpot mahjongpg soft link gamertp jackpotelemen sakti mahjongpola maxwin mahjongslot olympus mudah mainrtp live starlightrumus slot mahjongmahjong scatter hitamslot pragmaticjam gacor mahjongpola gacor mahjongstrategi maxwin olympusslot jamin menangrtp slot gacorscatter wild banditopola slot mahjongstrategi maxwin sweet bonanzartp slot terakuratkejutan scatter hitamslot88 resmimaxwin olympuspola mahjong pgsoftretas mahjong waystrik mahjongtrik slot olympusewallet modal recehpanduan pemula slotpg soft primadona slottercheat mahjong androidtips dewa slot mahjongslot demo mahjonghujan scatter olympusrtp caishen winsrtp sweet bonanzamahjong vs qilinmaxwin x5000 starlight princessmahjong wins x1000rtp baru wild scatterpg soft trik maxwinamantotorm1131