Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2015

Open Access 01.12.2015 | Research

Tumor progression locus 2 ablation suppressed hepatocellular carcinoma development by inhibiting hepatic inflammation and steatosis in mice

verfasst von: Xinli Li, Chun Liu, Blanche C. Ip, Kang-Quan Hu, Donald E. Smith, Andrew S. Greenberg, Xiang-Dong Wang

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2015

Abstract

Background

Tumor progression locus 2 (TPL2), a serine-threonine kinase, functions as a critical regulator of inflammatory pathways and mediates oncogenic events. The potential role of Tpl2 in nonalcoholic fatty liver disease (NAFLD) associated hepatocellular carcinoma (HCC) development remains unknown.

Methods

Both wild-type and Tpl2 knockout male mice were initiated by a hepatic carcinogen (diethylnitrosamine, i.p. with a single dose of 25 mg.kg−1)at 2 weeks of age, and then were given the high carbohydrate diet feeding to induce hepatic steatosis, inflammation, adenoma and HCC for 24 weeks.

Results

Tpl2 knockout mice had significantly lower incidences of liver tumor and developed hepatocellular adenoma only, which is contrast to wild-type mice where they all developed HCC. Tpl2 knockout mice had significantly down-regulated phosphorylation of JNK and ERK, and levels of mRNA expression of pro-inflammatory cytokines (Il-1β, Il-18, Mcp-1 and Nalp3), which correlated with the reduced incidence and number of hepatic inflammatory foci. Furthermore, Tpl2 ablation resulted in decreased hepatic steatosis and expression of de novo lipogenesis related markers (ACC, SCD1, SREBP1C and AKT phosphorylation), as well as reduction of endoplasmic reticulum stress biomarkers PERK and eIF-2a.

Conclusion

The study revealed for the first time that Tpl2 plays a significant role in promoting HCC development by its pro-inflammatory effect, which suggested that Tpl2 could be a molecular target for HCC prevention.
Hinweise

Competing interests

The authors declare no conflicts of interest.

Authors’ contributions

XLL and KQH conducted RT-PCR and Western blot. CL participated in the histo-pathological analysis. DES and ASG participated in the animal breeding. BCI and XDW participated in the design of the study. XLL performed the statistical analysis. XLL and XDW drafted the manuscript. All authors read and approved the final manuscript.
Abkürzungen
HCC
hepatocellular carcinoma
NAFLD
non-alcoholic fatty liver disease
TG
triglycerides
NASH
nonalcoholic steatohepatitis
TPL2
tumor progression locus 2
MAP3K8
MAP3 kinase 8
ERKs
extracellular signal regulated kinases
JNKs
c-jun N-terminal kinases
TNF-α
tumor necrosis factor-α
IL-1β
interleukin-1β
HFD
high fat diet
DEN
diethylnitrosamine
HCD
high carbohydrates diet
AKT
protein kinase B
ER
endoplasmicreticulum
PERK
PKR-like kinase
ACC
acetyl-CoA carboxylase
SCD
stearoyl-CoA desaturase
p-
phosphorylated
t-
total
GRP78
Glucose regulated protein 78
eIF2a
eukaryotic translation initiation factor 2a
SREBP-1c
sterol regulatory element-binding protein-1c
CHOP
C/EBP homologues protein
SD
standard deviation
SEM
standard error of the mean
IL-18
Interleukin-18
MCP-1
monocyte chemotactic protein 1
NALP3
NACHT, LRR and PYD domains-containing protein 3
DNL
de novo lipogenesis

Background

Hepatocellular carcinoma (HCC) is the sixth most prevalent human malignancy in the world and the third leading cause of cancer-related mortality [1]. Relative 5-year survival rate of HCC is only 15 %, which emphasizes the importance of primary prevention of HCC. Chronic infection by hepatitis B and hepatitis C virus, exposure to aflatoxin, alcoholic injury and genetic disorders have proven to play a critical role in the development of HCC [2], however, the etiology remains unknown in almost 50 % of HCC patients. Recent studies suggest that nonalcoholic fatty liver disease (NAFLD) is associated with an increased risk of HCC [3, 4], but it remains unclear whether NAFLD is a causative factor for HCC [5].
Steatosis is the initial stage of NAFLD, which can progress into more pathological stages including nonalcoholic steatohepatitis (NASH), fibrosis and cirrhosis, with the result of the increased risk for HCC development. Previous studies had indicated the significant contribution of NASH to HCC development, where pro-inflammatory cytokine and chemokines favors malignant transformation of hepatocytes by providing a tumor microenvironment [6, 7]. Inflammatory cascades through interactions of numerous signaling pathways progressively stimulated hepatocyte proliferation and apoptosis [8]. However, the potential causal relationships among inflammation, steatosis and HCC development need more supporting evidence.
Tumor progression locus 2 (TPL2), a serine-threonine kinase, functions as a critical regulator of inflammatory pathways and mediates oncogenic events by phosphorylating its downstream targets extracellular signal regulated kinases (ERKs), c-Jun N-terminal kinases (JNKs) and P38 [9] and subsequently up-regulating the production of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) [10]. The role of Tpl2 in acute and chronic inflammatory disorders and the determination of cellular death/survival ratios in the inflammatory microenvironment had been well-documented [11]. Our previous work demonstrated that whole-body ablation of Tpl2 attenuates high fat diet (HFD) induced hepatic inflammatory lesions compared to wild-type control mice, with a concomitant significant reduction in hepatic inflammatory genes expression [12]. However, the potential role of Tpl2 in tumorigenesis remains inconsistent [11, 13]. Several reports supported its oncogenic role in breast cancer [14], lymphoma [15] and prostate cancer [16], while others had suggested an anti-oncogenic activity in lung [17], colitis-associated tumorigenesis [18] and skin tumorigenesis [19]. This inconsistency could be attributed to the complexity of Tpl2’s role in terms of specific organs, different stages of carcinogenesis or the animal models used. However, there had no reports investigating the potential contribution of Tpl2 to HCC development by using Tpl2 knockout mouse model to date.
In the present study, we investigate the role of Tpl2 and its potential mechanisms in the development of hepatic steatosis, inflammation and tumors including HCC. Both wild-type and Tpl2 knockout male mice were initiated with [diethylnitrosamine (DEN)] at 2 weeks of age, and 4 weeks later, both groups mice were given the high carbohydrate diet (HCD) feeding for 24 weeks to induce hepatic steatosis, inflammation and HCC.

Methods

Animals, diet, carcinogen and study design

Tpl2 knockout mice were provided by Dr. Philip Tschilis (Tufts University) and backcrossed into C57BL/6J mice for >10 generations, as previously described [12, 20]. Genotype of animals was verified at weaning and again before the animals were killed at the conclusion of the experiment. Tpl2 mRNA expression was detected in wild-type but not in Tpl2 knockout mice (data not shown). All male C57BL/6J wild-type mice and Tpl2 knockout mice were injected i.p. with a single dose of 25 mg.kg−1 BW filter-sterilized, > 99.9 % purity DEN (Sigma-Aldrich, St. Louis, MO) at 2 weeks after birth, as previously described [21]. At 6 weeks of age, both wild-type (n = 26) and Tpl2 knockout (n = 20) mice were housed individually and fed a powdered, high carbohydrate, low fat diet [HCD, 12 % fat, 22 % protein, 66 % carbohydrates based on total caloric content (Bio-Serv, Flemington, NJ)] ad libitum for 24 weeks, as previously described [22]. Mice were killed by terminal exsanguination under deep isoflurane anesthesia followed by vital organ removal. All animal protocols and procedures were conducted under the approval of the Institutional Animal Care and Use Committee at the Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University. All animals received human care and that study protocols comply with the institution's guidelines.

Histopathology procedures and evaluation

Briefly, two investigators, blinded to the treatment groups, identified/counted the liver tumors (tumor incidence and multiplicity) on the surface of liver. The left lobe of the mouse liver was fixed in 10 % buffered formalin solution (Thermo Fisher Scientific, Waltham MA), processed and embedded in paraffin for serial sectioning for pathological analysis. The remaining lobes of the liver were divided into smaller portions, snap-frozen in liquid nitrogen and stored at -80 °C.
Five-micrometer sections of liver tissue were stained with hematoxylin (H) and eosin (E) (H&E) for histopathologic examination. Two independent investigators, blinded to treatment groups, examined the sections under light microscopy and identified hepatic lesions (including hyperplasia, hepatocellular adenoma and HCC). Liver histopathology of non-tumor areas was graded for steatosis magnitude (both macro- and micro- vesicular) and liver inflammation severity (inflammatory foci) as previously described [21]. Briefly, the degree of steatosis was graded 0–3 [grading 0: <5 % (normal), 1: 5 %-25 %, 2: 26 %-50 %, 3: >51 %] based on the average percent of fat-accumulated hepatocytes per field at 100 × magnification in 20 random fields. Inflammatory foci were evaluated by the number of inflammatory cell clusters (mononuclear inflammatory cells) in 20 random fields at 100 × magnification. The twenty fields of view at 100 × magnification represented 0.63 cm2 and inflammatory foci counts were represented as the number of foci per cm2. A ZEISS microscope with a PixeLINK USB 2.0 (PL-B623CU) digital Camera and PixeLINK μScope Microscopy Software (Ottawa ON, Canada) was used for image capture for all histological analyses.

RNA extraction and real time-PCR

RNA of liver tissue was isolated using TriPure Isolation Reagent (Roche, NJ) according to the manufacturer’s instructions. cDNA was synthesized using M-MLV reverse transcriptase (Invitrogen, Grand Island, NY). Real-time PCR reactions were carried out using SYBR green (Fast Start Universal SYBR Green Master, Roche). Relative changes in gene expression were determined using the 2-ΔΔCt method and normalized to the control of actin.

Western blot analysis

Hepatic whole cell lysate protein was extracted utilizing previously described methods [23]. Protein concentrations of the sample were assessed using the Bradford assay (Bio-Rad, Hercules CA). The protein expression bands were quantified using a densitometer (GS-710 calibrated imaging densitometer, Bio-Rad). The antibodies against acetyl-CoA carboxylase (ACC), stearoyl-CoA desaturase 1 (SCD-1), total- and phosphor- AKT (p-AKT, t-AKT), glucose regulated protein 78 (GRP78/Bip), total- and phosphor- (Thr980) PERK (t-PERK, p-PERK), total- and phosphor- (Ser51) eukaryotic translation initiation factor 2a (t-eIF2a, p-eIF2a), total- and phosphor- (Thr183/Tyr185) JNK (p-JNK, t-JNK), total- and phosphor- ERK (p-ERK, t-ERK) were all purchased from Cell Signaling (Danvers, MA). The antibodies against sterol regulatory element-binding protein-1c (SREBP-1c) and C/EBP homologues protein (CHOP) were purchased from Santa Cruz Biotechnology (Dallas, TX).

Statistical analyses

Data are presented as mean ± standard deviation (SD) for animal body weights, liver tumor numbers per animal and inflammatory foci, and nonparametric test was performed except for the comparison of the final body weights which was conducted using an independent t-test. The incidence of hepatic lesions and inflammation foci was compared by Chi-squares test. Steatosis grading was presented as median (grading range) and a non-parametric test was performed. Data are presented as mean ± standard error of the mean (SEM) for mRNA and protein levels, and t-test was conducted. SPSS software was used for all statistical analysis, and P < 0.05 indicated the significant difference when comparing between wild-type and Tpl2 knockout mice.

Results

Effect of Tpl2 ablation on body weight, liver weight and hepatic tumorigenesis

Tpl2 knockout mice had significantly lower liver weight and final body weight than those of wild-type control, although there had no significant difference in the ratio of liver weight/body weight between two groups (Table 1). The tumor incidence on the surface of liver in Tpl2 knockout mice was significantly lower than wild-type mice (75 % vs 100 % respectively, P < 0.05, Table 1). The number of hepatic tumor had no statistical difference between those two groups. The pathological analysis demonstrated that all wild-type mice (26 out of total 26 mice) developed hyperplasia, hepatocellular adenoma (Fig. 1c, 1d) and HCC (Fig. 1e, 1f) after the 24-week HCD feeding (Table 1). In contrast, the Tpl2 knockout mice with positive tumor on the surface of the liver (15 out of 20) developed only hyperplasia and hepatocellular adenoma, and no HCC detected (Table 1).
Table 1
Study outcomes
Index
Wild type group
Tpl2 knockout group
Animal number (n)
26
20
Final body weight (g)
41.2 ± 3.2
38.1 ± 5.9*
Liver weight (g)
2.5 ± 0.8
2.1 ± 0.3*
Liver/body weights (%)
6.2 ± 2.4
5.6 ± 1.1
Incidence of liver tumor (%)
100 % (26/26)
75 % (15/20)*
Liver tumor number/per animal
11 ± 6
10 ± 5
Histopathology of hepatic lesion#
n
%
n
%
Hyperplasia
26
100
3
15*
Hepatocellular adenoma
26
100
12
60*
Hepatocellular carcinoma
26
100
0
0*
Incidence of inflammation foci (%)
65.4% (17/26)
20 % (4/20)*
Inflammation foci number (cm2)
2.2 ± 2.4
0.6 ± 1.4*
Hepatic steatosis (median)
2
1*
Hepatic steatosis grading
n
%
n
%
 0
1
3.8
8
40
 1
2
7.7
4
20
 2
15
57.7
4
20
 3
8
30.8
4
20
#Each animal has more than one type of lesions
Values are expressed as means ± SD. An Independent t-test was performed except the comparison of incidence of liver tumor and inflammatory foci between WT and Tpl2 KO mice, which is conducted by nonparametric test. For steatosis, 20 images at 100 × magnification were captured for each section and blindly evaluated twice to determine grade of steatosis (both macro- and micro-vesicular) by two blinded investigators. The degree of steatosis was graded 0-3 based on the area of the liver section occupied by fat vacuoles. Data are presented as median (grading range) and Nonparametric test was used to test for statistical significance between groups for ordinal variable (liver steatosis score)
For each given row, * indicates a significant difference between groups (P < 0.05)

Tpl2 ablation decreased hepatic inflammatory responses and suppressed the activation of JNK and ERK signaling molecules

Hepatic inflammatory foci was detected in 65 % of the wild-type mice but only in 20 % of the Tpl2 knockout mice (P < 0.05; Table 1, Fig. 1b). Tpl2 knockout mice had significantly less hepatic inflammatory foci in contrast to wild-type mice (0.6 vs 2.2, P < 0.05; Table 1), accompanying with lower mRNA level of genes [Il-1β, interleukin-18 (Il-18), monocyte chemotactic protein 1 (Mcp1), NACHT, LRR and PYD domains-containing protein 3 (Nalp3) related to hepatic inflammation (Fig. 2a)]. In addition, there was a significant decrease in the phosphorylation of JNK1/2 and ERK1/2, the downstream targets of TPL2-mediated inflammation signaling, in Tpl2 knockout mice as compared with wild-type mice (Fig. 2b).

Tpl2 ablation alleviated hepatic steatosis, and down-regulated protein and mRNA expression of molecules involved in de novo lipogenesis (DNL) and endoplasmic reticulum (ER) stress

Tpl2 knockout mice had lower steatosis grades compared to wild-type mice (Table 1). Only one of 26 wild-type mice (3.8 %) had a steatosis grading of 0 compared to 8 out of 20 Tpl2 knockout mice (40 %). In contrast, 15 out of 26 wild-type mice (57.7 %) developed steatosis grade of 2 as compared to 4 out of 20 Tpl2 knockout mice (20 %). Steatosis grades were statistically different between the 2 groups (medians of 2 vs. 1 for wild-type vs. Tpl2 knockout, respectively, P < 0.05, Table 1). In contrast to wild-type mice, Tpl2 knockout mice had significantly decreased proteins expression of ACC and SCD1, two lipogenic proteins (Fig. 3a), and decreased protein expression of SREBP1C (Fig. 3a), which is one of the transcription factors that regulates expression of genes involved in DNL. Meanwhile, we had detected the decreased expression of phosphorylated AKT (Fig. 3b), which is consistent with the decreased SREBP1c expression (Fig. 3a) and hepatic steatosis (Table 1) in Tpl2 knockout mice.
Increased ER stress can promote HCC development and progression by activating the fibrogenic activity of hepatic stellate cells with subsequent liver cirrhosis [24]. In the present study, Tpl2 knockout mice had decreased hepatic PERK and eIF2α phosphorylation compared to wild-type mice (Fig. 4), but Tpl2 ablation did not alter the expression of the chaperon factor GRP78 that attenuates ER stress or PERK mediated pro-apoptotic protein CHOP (data not shown).

Discussion

The role of hepatic inflammation induced by dietary factors, such as HCD and HFD, in promoting DEN-initiated HCC development had been demonstrated in previous studies [6, 22, 25]. The present study, for the first time, revealed the significantly lower incidence of hepatic tumor with no HCC development in HCD-fed, Tpl2 knockout mice in contrast to wild-type mice which all developed HCC. The significant difference in tumor pathological types between Tpl2 knockout mice and wild-type mice supported the critical role of Tpl2 as a promoter in tumor progression from hepatic hepatocellular adenoma to HCC. Furthermore, we provided a strong evidence that the TPL2 ablation decreased hepatic inflammatory response and hepatic steatosis in Tpl2 knockout mice. These effects could inhibit the malignant transformation of hepatocytes and the progression of liver tumor by suppressing tumor-promoting microenvironment and alleviating the malignant effects of dys-regulation of lipid metabolism on HCC.
TPL2 mediated inflammatory response by phosphorylating ERK and JNK, two downstream targets of TPL2 signaling pathway. In our study, the significant lower protein levels of hepatic p-JNK and p-ERK in Tpl2 knock out mice was consistent with the decreased hepatic inflammation, which supported the role of activated ERK (p-ERK) and JNK (p-JNK) in mediating the pro-inflammatory effect of Tpl2. The decreased hepatic inflammation induced by HCD feeding supported by the fewer hepatic inflammatory foci detected and the lower levels of inflammatory cytokine expression of Il-1β, Il-18, Mcp-1 and Nalp3 in Tpl2 knockout mice, as compared with wide type mice. This is also in agreement with our previous study that Tpl2 knockout mice fed HFD had lower levels of inflammation compared to wild-type mice [12]. The present work further indicated the role of Tpl2 in mediating hepatic inflammation and HCC development induced by HCD.
Elevated hepatic DNL could promote hepatic steatosis [26]. Interestingly, in our present study, Tpl2 ablation resulted in a significant decrease of hepatic steatosis, and the down-regulated protein expression of genes related to DNL, such as ACC and SCD1, which was associated the decreased protein expression of AKT phosphorylation and SREBP-1C.
Previous studies have suggested that AKT activation is essential and sufficient to stimulate DNL and lipid accumulation through the induction of SREBP-1C [27, 28]. Furthermore, the promoting role of dysregulated lipid metabolism [29] and lipogenesis induced by activated AKT in HCC development had been documented [30]. Thus, the decrease in steatosis and AKT activation could further explain the decreased HCC in Tpl2 knockout mice.
However, the exact role of Tpl2 in regulating the genes related to DNL is unclear. It has been reported that the regulation of lipid metabolism and hepatic steatosis mediated by the activation of JNK, the downstream target of TPL2, is associated with activated ER stress, especially in the PERK-eIF2a pathway [31]. Our observation of the down-regulated expression of p-JNK, PERK and p-eIF2a in Tpl2 knockout mice with the decreased hepatic steatosis supported the involvement of ER stress in TPL2/JNK mediated steatosis. Recent reports have demonstrated that ER stress is closely associated with hepatic lipogenesis with elevated DNL [26, 32, 33], and population-based studies also support the positive regulation of ER stress in hepatic lipogenesis [34, 35]. Since newly synthesized unfolded proteins in the ER is a major cause of activated ER stress and activated ER stress could further induce lipogenesis, the vicious cycle could result in the progression of steatosis [21]. In our present study, the deceased expression of the lipogenic enzymes ACC, SCD1 and SREBP-1C could decrease the impact of protein folding in the ER, and alleviate ER stress in Tpl2 knockout mice. PERK-mediated signaling can also promote apoptosis through inducing pro-apoptotic CHOP expression, the no significant difference of the protein level of CHOP suggested that HCD induced expression of PERK predominantly mediated lipogenesis, but not pro-apoptotic effects in our present study. Furthermore, the expression of GRP78, a chaperone protein that attenuates ER stress [36], were not different between the Tpl2 knockout and wild-type mice, combining with our observation that Tpl2 knockout mice had relatively lower levels of PERK and p-eIF2a, the downstream molecular of GRP78, than wild-type mice, we therefore concluded that Tpl2 mediated hepatic lipogenesis by targeting the axis of TPL2/JNK/ER stress/p-eIF2a, the downstream of GRP78.
Our previous study had shown that HCD feeding promoted DEN-initiated HCC development accompanying with the induction of the hepatic ER stress-mediated PERK activation, which subsequently induced the elevated expression of pro-survival markers AKT and ERK1/2 [22]. Our present study demonstrated that Tpl2 ablation decreased ER stress mediated PERK expression and eIF-2a activation, which might account for the decreased tumor incidence. Since ER-dependent cell fate is associated with the activation of JNK/ERK [37], and the pro-survival role of activated ERK can be mediated by AKT phosphorylation and the involvement of PERK/eIF2a signaling [3840], thus, in the present study, the down-regulated activation of ERK, AKT and PERK/eIF2a provided further explanation for the decreased incidence of HCC in Tpl2 knockout mice. The exact relationship between TPL2 and ER stress requires further investigation.
In summary, our present study demonstrated that Tpl2 played significant role in DEN-initiated, NAFLD associated HCC development by using Tpl2 knockout mouse model. Both TPL2/ERK/JNK axis mediated hepatic inflammation and TPL2/JNK/ERS/p-eIF2a axis mediated hepatic lipogenesis synergistically promoted HCC development. These data provided strong molecular evidence supporting Tpl2 as a promoter in HCC development. Interestingly, it has been shown that luteolin, one of the common phytonutrients present in celery, parsley, broccoli and herbal spices, could target Tpl2 and inhibit its activity in vitro [41]. Recent in vivo studies also supported the preventive effects of luteolin on DEN-initiated alcohol-promoted hepatic carcinogenesis in mice [42], and DEN-initiated HCC development in rats [43]. Taking all into consideration, dietary or pharmacologic interventions targeting Tpl2 could be a potential direction for HCC prevention in the future.

Conclusions

This is the first study to report that tumor progression locus 2 (Tpl2) plays a significant role in promoting nonalcoholic fatty liver disease and hepatocellular carcinoma (HCC) development. Targeting Tpl2 might be a potential direction for HCC prevention.

Acknowledgments

The author thanks Dr. Camilla P. Stice, and the HNRCA Cancer Cluster members for their contributions to this study. Dr. Xinli Li was supported by a scholarship (File NO. 2011832272) from the State Scholarship Fund by The China Scholarship Council.
Financial support was provided by the NIH grant (CA176256), USDA/ARS grant (58-1950-0014), and HNRCA Cancer Cluster funding.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare no conflicts of interest.

Authors’ contributions

XLL and KQH conducted RT-PCR and Western blot. CL participated in the histo-pathological analysis. DES and ASG participated in the animal breeding. BCI and XDW participated in the design of the study. XLL performed the statistical analysis. XLL and XDW drafted the manuscript. All authors read and approved the final manuscript.
Literatur
3.
Zurück zum Zitat Mittal S, Sada YH, El-Serag HB, Kanwal F, Duan Z, Temple S, et al. Temporal Trends of Nonalcoholic Fatty Liver Disease-Related Hepatocellular Carcinoma in the Veteran Affairs Population. Clin Gastroenterol Hepatol. 2014. doi:10.1016/j.cgh.2014.08.013.PubMedCentral Mittal S, Sada YH, El-Serag HB, Kanwal F, Duan Z, Temple S, et al. Temporal Trends of Nonalcoholic Fatty Liver Disease-Related Hepatocellular Carcinoma in the Veteran Affairs Population. Clin Gastroenterol Hepatol. 2014. doi:10.​1016/​j.​cgh.​2014.​08.​013.PubMedCentral
8.
Zurück zum Zitat Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113–21.CrossRefPubMed Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113–21.CrossRefPubMed
9.
Zurück zum Zitat Aoki M, Akiyama T, Miyoshi J, Toyoshima K. Identification and characterization of protein products of the cot oncogene with serine kinase activity. Oncogene. 1991;6(9):1515–9.PubMed Aoki M, Akiyama T, Miyoshi J, Toyoshima K. Identification and characterization of protein products of the cot oncogene with serine kinase activity. Oncogene. 1991;6(9):1515–9.PubMed
14.
16.
Zurück zum Zitat Lee HW, Cho HJ, Lee SJ, Song HJ, Park MC, Seol HJ, et al. Tpl2 induces castration resistant prostate cancer progression and metastasis. Int J Cancer. 2014. doi:10.1002/ijc.29248. Lee HW, Cho HJ, Lee SJ, Song HJ, Park MC, Seol HJ, et al. Tpl2 induces castration resistant prostate cancer progression and metastasis. Int J Cancer. 2014. doi:10.​1002/​ijc.​29248.
20.
Zurück zum Zitat Dumitru CD, Ceci JD, Tsatsanis C, Kontoyiannis D, Stamatakis K, Lin JH, et al. TNF-alpha induction by LPS is regulated posttranscriptionally via a Tpl2/ERK-dependent pathway. Cell. 2000;103(7):1071–83.CrossRefPubMed Dumitru CD, Ceci JD, Tsatsanis C, Kontoyiannis D, Stamatakis K, Lin JH, et al. TNF-alpha induction by LPS is regulated posttranscriptionally via a Tpl2/ERK-dependent pathway. Cell. 2000;103(7):1071–83.CrossRefPubMed
21.
Zurück zum Zitat Ip BC, Hu KQ, Liu C, Smith DE, Obin MS, Ausman LM, et al. Lycopene metabolite, apo-10'-lycopenoic acid, inhibits diethylnitrosamine-initiated, high fat diet-promoted hepatic inflammation and tumorigenesis in mice. Cancer Prev Res (Phila). 2013;6(12):1304–16. doi:10.1158/1940-6207.capr-13-0178.CrossRef Ip BC, Hu KQ, Liu C, Smith DE, Obin MS, Ausman LM, et al. Lycopene metabolite, apo-10'-lycopenoic acid, inhibits diethylnitrosamine-initiated, high fat diet-promoted hepatic inflammation and tumorigenesis in mice. Cancer Prev Res (Phila). 2013;6(12):1304–16. doi:10.​1158/​1940-6207.​capr-13-0178.CrossRef
28.
Zurück zum Zitat Fleischmann M, Iynedjian PB. Regulation of sterol regulatory-element binding protein 1 gene expression in liver: role of insulin and protein kinase B/cAkt. Biochem J. 2000;349(Pt 1):13–7.PubMedCentralCrossRefPubMed Fleischmann M, Iynedjian PB. Regulation of sterol regulatory-element binding protein 1 gene expression in liver: role of insulin and protein kinase B/cAkt. Biochem J. 2000;349(Pt 1):13–7.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Nakanishi K, Sakamoto M, Yamasaki S, Todo S, Hirohashi S. Akt phosphorylation is a risk factor for early disease recurrence and poor prognosis in hepatocellular carcinoma. Cancer. 2005;103(2):307–12. doi:10.1002/cncr.20774.CrossRefPubMed Nakanishi K, Sakamoto M, Yamasaki S, Todo S, Hirohashi S. Akt phosphorylation is a risk factor for early disease recurrence and poor prognosis in hepatocellular carcinoma. Cancer. 2005;103(2):307–12. doi:10.​1002/​cncr.​20774.CrossRefPubMed
34.
Zurück zum Zitat Boden G, Duan X, Homko C, Molina EJ, Song W, Perez O, et al. Increase in endoplasmic reticulum stress-related proteins and genes in adipose tissue of obese, insulin-resistant individuals. Diabetes. 2008;57(9):2438–44. doi:10.2337/db08-0604.PubMedCentralCrossRefPubMed Boden G, Duan X, Homko C, Molina EJ, Song W, Perez O, et al. Increase in endoplasmic reticulum stress-related proteins and genes in adipose tissue of obese, insulin-resistant individuals. Diabetes. 2008;57(9):2438–44. doi:10.​2337/​db08-0604.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Das SK, Chu WS, Mondal AK, Sharma NK, Kern PA, Rasouli N, et al. Effect of pioglitazone treatment on endoplasmic reticulum stress response in human adipose and in palmitate-induced stress in human liver and adipose cell lines. Am J Physiol Endocrinol Metab. 2008;295(2):E393–400. doi:10.1152/ajpendo.90355.2008.PubMedCentralCrossRefPubMed Das SK, Chu WS, Mondal AK, Sharma NK, Kern PA, Rasouli N, et al. Effect of pioglitazone treatment on endoplasmic reticulum stress response in human adipose and in palmitate-induced stress in human liver and adipose cell lines. Am J Physiol Endocrinol Metab. 2008;295(2):E393–400. doi:10.​1152/​ajpendo.​90355.​2008.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Oh SH, Lim SC. Endoplasmic reticulum stress-mediated autophagy/apoptosis induced by capsaicin (8-methyl-N-vanillyl-6-nonenamide) and dihydrocapsaicin is regulated by the extent of c-Jun NH2-terminal kinase/extracellular signal-regulated kinase activation in WI38 lung epithelial fibroblast cells. J Pharmacol Exp Ther. 2009;329(1):112–22. doi:10.1124/jpet.108.144113.CrossRefPubMed Oh SH, Lim SC. Endoplasmic reticulum stress-mediated autophagy/apoptosis induced by capsaicin (8-methyl-N-vanillyl-6-nonenamide) and dihydrocapsaicin is regulated by the extent of c-Jun NH2-terminal kinase/extracellular signal-regulated kinase activation in WI38 lung epithelial fibroblast cells. J Pharmacol Exp Ther. 2009;329(1):112–22. doi:10.​1124/​jpet.​108.​144113.CrossRefPubMed
39.
Zurück zum Zitat Koumenis C, Naczki C, Koritzinsky M, Rastani S, Diehl A, Sonenberg N, et al. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha. Mol Cell Biol. 2002;22(21):7405–16.PubMedCentralCrossRefPubMed Koumenis C, Naczki C, Koritzinsky M, Rastani S, Diehl A, Sonenberg N, et al. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha. Mol Cell Biol. 2002;22(21):7405–16.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Mounir Z, Krishnamoorthy JL, Wang S, Papadopoulou B, Campbell S, Muller WJ et al. Akt determines cell fate through inhibition of the PERK-eIF2alpha phosphorylation pathway. Sci Signal. 2011;4(192):ra62. doi:10.1126/scisignal.2001630. Mounir Z, Krishnamoorthy JL, Wang S, Papadopoulou B, Campbell S, Muller WJ et al. Akt determines cell fate through inhibition of the PERK-eIF2alpha phosphorylation pathway. Sci Signal. 2011;4(192):ra62. doi:10.​1126/​scisignal.​2001630.
41.
Zurück zum Zitat Kim JE, Son JE, Jang YJ, Lee DE, Kang NJ, Jung SK, et al. Luteolin, a novel natural inhibitor of tumor progression locus 2 serine/threonine kinase, inhibits tumor necrosis factor-alpha-induced cyclooxygenase-2 expression in JB6 mouse epidermis cells. J Pharmacol Exp Ther. 2011;338(3):1013–22. doi:10.1124/jpet.111.179200.CrossRefPubMed Kim JE, Son JE, Jang YJ, Lee DE, Kang NJ, Jung SK, et al. Luteolin, a novel natural inhibitor of tumor progression locus 2 serine/threonine kinase, inhibits tumor necrosis factor-alpha-induced cyclooxygenase-2 expression in JB6 mouse epidermis cells. J Pharmacol Exp Ther. 2011;338(3):1013–22. doi:10.​1124/​jpet.​111.​179200.CrossRefPubMed
42.
Zurück zum Zitat Rafacho CPS BPM, Liu C, Greenberg AS, Ausman LM, Wang XD. Inhibition of diethylnitrosamine-initiated alcohol-promoted hepatic inflammation and precancerous lesions by flavonoid luteolin is associated with increased sirtuin 1 activity in mice. HepatoBiliary Sur Nutr. 2015;4:124–34. Rafacho CPS BPM, Liu C, Greenberg AS, Ausman LM, Wang XD. Inhibition of diethylnitrosamine-initiated alcohol-promoted hepatic inflammation and precancerous lesions by flavonoid luteolin is associated with increased sirtuin 1 activity in mice. HepatoBiliary Sur Nutr. 2015;4:124–34.
Metadaten
Titel
Tumor progression locus 2 ablation suppressed hepatocellular carcinoma development by inhibiting hepatic inflammation and steatosis in mice
verfasst von
Xinli Li
Chun Liu
Blanche C. Ip
Kang-Quan Hu
Donald E. Smith
Andrew S. Greenberg
Xiang-Dong Wang
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2015
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-015-0254-2

Weitere Artikel der Ausgabe 1/2015

Journal of Experimental & Clinical Cancer Research 1/2015 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.