2023 Impact Factor
Leptin, a hormone predominantly derived from adipose tissue, has long been considered to play regulatory roles in food intake and energy expenditure, since the discovery of obese gene (Zhang
IL-1β signaling plays important roles in the development and/or progression of many inflammation-associated diseases, such as arthritis and diabetes mellitus (Mirea
Gene expression and maturation of IL-1β is tightly regulated at multiple steps. IL-1β is initially synthesized as an inactive precursor and converted into a biologically active form through proteolytic cleavages (Martinon
Cell cycle, a series of events involved in cell growth and division, is regulated by a number of regulatory proteins in a concerted action. Indeed, a group of cyclins and cyclin-dependent kinases (CDKs) promote the progression of the cycle, while various CDK inhibitors, including p53, p21, p16, and retinoblastoma, act as negative regulators of the cell cycle (Kumari and Jat, 2021). It has been well documented that leptin induces cell cycle progression in cancer cells via up-regulation of cyclin D1 (Chen
Leptin exerts a direct cytotoxic effect in hepatocytes, which may be critical in the development of pathological conditions in the liver. In the present study, to better understand the mechanisms underlying leptin-induced hepatic damage, we elucidated the molecular mechanisms by which leptin induces cytotoxic effects in hepatocytes, particularly focusing on the role of IL-1β signaling. We have demonstrated that maturation and secretion of IL-1β critically contributes to the apoptotic cell death and cell cycle arrest by leptin in rat hepatocytes. In addition, cell death and cell cycle arrest by IL-1β signaling are mediated, at least in part, via activation of p38MAPK and suppression of AKT signaling.
Type IV collagenase (C5138), type I collagen (C3867), Ac-YVAD-cmk (SML0429), a pharmacological inhibitor of caspase-1, Hank’s Balanced Salt Solution (HBSS) (H4891), and recombinant leptin (L3772) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Propidium iodide dye (ab139418), MitoOrange dye (ab138898), and antibodies against cytokeratin-18 (ab181597) and p16 (ab51243) were procured from Abcam (Cambridge, UK). SC79 (HY-18749) were obtained from MedChem Express (Monmouth Junction, NJ, USA). Annexin V & 7-AAD assay kit (640922) was purchased from BioLegend (San Diego, CA, USA). Primary antibodies against Bax (2772), Bcl-2 (2876), pro-caspase-3 (9662), cleaved caspase-3 (9664), PARP (9542), phosphor-AKT (4060S), phosphor-GSK3β (5558S), and phosphor-p38MAPK (9215) were acquired from Cell Signaling Technologies (Danvers, CO, USA). IL-1β (MAB501) and IgG1 isotype control (MAB002) antibodies were bought from R&D Systems (Minneapolis, MN, USA). Anti-rabbit (31460) and anti-mouse (31430) horseradish-conjugated secondary antibodies were purchased from Thermo Scientific (Waltham, MA, USA).
Hepatocytes were isolated from 6-7 weeks old male Sprague Dawley (SD) rats using a two-step collagenase perfusion process as described previously (Baral and Park, 2021). All animal experiments were performed as per the guidelines of Yeungnam University Institutional Animal Care and Use of Committee (IACUC). The experimental protocols were reviewed and approved by the Yeungnam University IACUC (Approval number: 2022-016). Briefly, after cannulation of hepatic portal vein using an 18G catheter, the liver was perfused with HBSS free of Ca2+ and Mg2+, and type IV collagenase (0.05%). Collagenase perfusion was initiated once the liver turned pale showing clear signs of RBC removal. After digestion of the liver, cells were collected by filtering through a cell strainer (100 µm) and washed twice with HBSS. Cells were resuspended in William’s Medium E containing 10% fetal bovine serum and 1% penicillin/streptomycin and seeded in collagen-coated dishes. After 2 h, the media were changed to remove the unattached cells.
IL-1R1loxP/loxP mice (# 028398) and mice expressing Cre transgene driven by the albumin promoter (AlbCre mice) (# 003574) on a C57BL/6 background were purchased from Jackson Laboratories (Farmington, CT, USA) and were housed under normal-pathogen free conditions. IL-1R1loxP/loxP and ALbCre mice were bred to obtain IL-1R1fl/fl: AlbCre (hereafter referred to as IL-1R1 KO mice). Similarly, age- and sex-matched littermates with IL-1R1fl/fl but not expressing Cre were used as wild type controls. Mice were then grouped based on their genetic backgrounds. Male mice aged between 6 to 8 weeks were used for the experiments.
Expression levels of IL-1R1 and Cre in WT and IL-1R1 KO mice were determined using PCR amplification. After incubating the tail piece in alkaline lysis buffer, the DNA samples were amplified using DreamTaq DNA polymerase system (Thermo Scientific). PCR products were subjected to gel electrophoresis (1.5-3% agarose gel), and images were captured using a Vilber Fusion Solo system (Vilber, Collegien, France).
Mice were administered leptin (1 mg/kg) twice daily for 15 days by intraperitoneal injection, whereas control group mice received an equivalent volume of 1X PBS via the same route. At the end of the treatment, animals were sacrificed by anesthetization, and the livers were excised and subjected for further experiments. For western blot analysis, the tissues were homogenized using radioimmunoprecipitation assay (RIPA) lysis buffer containing 1% protease inhibitors cocktail and lysed further using sonicator (Sonics & Materials Inc, Newtown, CT, USA).
Cell viability was measured based on the conversion of 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-
For measuring the expression levels of the genes of interest, Western blot analysis was carried out as described previously (Nguyen
Conditioned media were prepared as described previously (Pun
Apoptosis of hepatocytes were monitored using Annexin V Apoptosis Detection Kit (BioLegend) according to the manufacturer’s instruction. Briefly, after treatment as indicated, cells were detached from the culture plate, resuspended in Annexin V Binding Buffer, and incubated with staining buffer containing FITC-Annexin V and 7-AAD for 15 min in the dark at room temperature. Finally, apoptotic cells were detected by flow cytometry analysis using BD FACS Verse (BD Biosciences, Franklin lakes, NJ, USA).
Distribution of the cells in each cell cycle phase was monitored by propidium iodide (PI) staining and flow cytometry analysis as described previously (Pham
Mitochondrial membrane potential in hepatocytes was analyzed by staining with MitoOrange dye according to the manufacturer’s instructions. Briefly, cells were detached, incubated with MitoOrange Dye (2 µL, 200 ×) for 30 min at 37°C, and suspended in the assay buffer. Mitochondrial membrane potential was monitored by flow cytometry analysis.
For gene silencing of the gene of interest, hepatocytes were seeded in a collagen-coated 35-mm dish at a density of 5×105 cells/dish. After overnight culture, cells were transfected with a siRNA targeting the gene of interest or scrambled control siRNA using Lipofectamine RNAi MAX (Thermo Fisher, Rockford, IL, USA) according to the manufacturer’s instructions. Gene silencing efficiency was monitored by western blot analysis after 24 h after transfection. The oligonucleotide siRNAs were purchased from Bioneer (Daejon, Korea). The siRNA sequences used are presented in Table 1.
Table 1 Sequences of the nucleotides used for siRNA tranfection
Target gene | Forward sequence | Reverse sequence |
---|---|---|
IL-1R1 | 5'-CUAUGAUGCCUAUGUUCUU-3' | 5'-AAGAACAUAGGCAUCAUAG-3' |
P16 | 5'-GAUGAUGGGCAACGUCAAA-3' | 5'-CAACGCGAGACUAGCAUAU-3' |
Scrambled siRNA | 5'-CCUACGCCACCAAUUUCGU-3' | 5'-ACGAAAUUGGUGGCGUAGG-3' |
The mRNA levels of the target genes were determined by RT-qPCR as described previously (Pham and Park, 2022). In brief, hepatocytes were seeded in a collagen-coated 35-mm dish at a density of 1×106 cells/dish. Total cellular RNA was extracted using Qiazol lysis reagent (Qiagen, Hilden, Germany). Total RNA (1 µg) was used for the synthesis of complementary DNA (cDNA), which was subsequently amplified by a Light Cycler 1.5 system (Roche Diagnostics, Mannheim, Germany) using an absolute SYBR green capillary mix (Thermo Scientific). The sequences of the primers used in this study are listed in Table 2.
Table 2 Sequences of the primers used for PCR amplification
Target gene | Forward sequence | Reverse sequence |
---|---|---|
p16 | 5'-CGGTATCTACTCTCCTCCGC-3' | 5'-GTTGCCAGAAGTGAAGCCAA-3' |
p21 | 5'-AACTGGGGAGGGCTTTCTTT-3' | 5'-TGTCTTGTCTTCGCTGAGGT-3' |
p27 | 5'-AACTAACCCGGGACTTGGAG-3' | 5'-GAGACCCAATTGAAGGCACC-3' |
GAPDH | 5'-TGAACGGGGAAGCTCACTGG-3' | 5'-TCCACCACCCTGTTGCTGTA-3' |
Cyclin D1 | 5'-CCCCAACAACTTCCTCTCCT-3' | 5'-AGCTTCTTCCTCCACTTCCC-3' |
Cyclin E1 | 5'-AAGGAGGGTGCTACTTGACC-3' | 5'-TCAGCTGACAGTGGAGAAGG-3' |
Values are presented as mean ± standard error of the mean (SEM) from at least three independent experiments. Data were analyzed by One-Way-Analysis of Variance with Tukey’s multiple comparison test using GraphPad Prism version 5.0 (La Jolla, CA, USA). Groups were considered statistically significant if
We have previously shown the involvement of IL-1β signaling in cytotoxic effect of leptin in hepatocytes. In this study, we have further elucidated the molecular mechanisms by which IL-1β signaling mediates decreased viability of hepatocytes. To this end, we first confirmed the effect of leptin on activation and secretion of IL-1β in our experimental condition. As shown in Fig. 1A, leptin significantly enhances secretion of mature IL-1β in a time-dependent manner, as determined by western blot analysis using TCA-precipitates of the culture media, which is in line with the previous reports demonstrating the intracellular activation of IL-1β by leptin (Baral and Park, 2021). Given that cell viability is determined by the rate of cell death and proliferation, we first investigated whether leptin and IL-1β signaling modulate the cell cycle in hepatocytes. As shown in Fig. 1, leptin treatment induced significant increases in proportions of cells in the sub G1 and G1 phase, but decreased those in the S and G2 phase (Fig. 1B, 1C). Interestingly, pretreatment with interleukin-1 receptor antagonist (IL-1Ra) caused restoration of typical proportions in each cell cycle phase (Fig. 1B). Essentially similar results were observed by gene silencing of IL-1 receptor-1 (IL-1R1) (Fig. 1C), suggesting that leptin-induced cell cycle arrest is mediated via IL-1β signaling. To further characterize leptin-induced cell cycle arrest, we measured the expression levels of cell cycle regulators and observed that leptin significantly upregulates p16 expression at both mRNA and protein levels (Fig. 1D-1F), whereas no significant effects were observed in other cell cycle regulators, including p21, p27, cyclin D1, and cyclin E1 (Supplementary Fig. 1). In subsequent experiments to verify the functional role of p16 in leptin-modulation of the cell cycle, gene silencing of p16 restored changes in cell populations in each cell cycle phase (Fig. 1G). In addition, pretreatment with IL-1Ra notably inhibited leptin-induced p16 expression at both mRNA and protein levels (Fig. 1H, 1I, respectively), indicating that IL-1β signaling is implicated in leptin-induced upregulation of p16. Since IL-1β is activated through NLRP3 inflammasomes, we also investigated the role of NLRP3 inflammasomes and found that treatment with NLRP3 inflammasomes inhibitors (MCC950 and Ac-YVAD-cmk) almost completely abolished leptin-stimulated p16 expression (Fig. 1J), which are similar to the results from IL-1Ra pretreatment. Moreover, treatment with conditioned media obtained from leptin-treated hepatocytes cultures significantly increased p16 expression, which was restored in the presence of Ac-YVAD-cmk (Fig. 1K). Collectively, these results suggest that cell cycle arrest by leptin is mediated through IL-1β signaling, at least in part, via upregulation of p16.
To further characterize the decreased viability of hepatocytes caused by leptin, we examined whether IL-1β secretion also contributes to apoptotic cell death. As shown in Fig. 2, leptin treatment prominently enhanced apoptosis in hepatocytes, as evidenced by annexin V/7-AAD binding assay. Interestingly, treatment with IL-1Ra and gene silencing of IL-1R1 significantly alleviated leptin-induced apoptotic cell death (Fig. 2A, 2B, respectively). Additionally, neutralization of IL-1β by treatment with IL-1β monoclonal antibody caused restoration of leptin-decreased hepatocyte viability (Supplementary Fig. 2), collectively suggesting the critical role of IL-1β signaling in leptin-induced apoptosis of hepatocytes. The role of IL-1β signaling was further confirmed by measuring the levels of the genes related with apoptosis. As indicated in Fig. 2, leptin-induced cleavage of cytokeratin-18, a critical apoptotic marker in hepatocytes, was markedly prevented by pretreatment with IL-1Ra (Fig. 2C) and silencing of IL-1R1 (Fig. 2D). In addition, pretreatment with IL-1Ra also substantially suppressed leptin-induced increases in other conventional apoptotic markers, including cleaved PARP (Fig. 2E) and Bax (Fig. 2F), while recovered the suppressed Bcl-2 expression (Fig. 2G). Furthermore, treatment with conditioned media obtained from hepatocyte culture in the presence of Ac-YVAD-cmk showed a marked reduction in leptin-stimulated caspase-3 cleavage (Fig. 2H). Finally, leptin-induced reduction in mitochondrial membrane potential, which can promote the release of apoptosis promoters such as cytochrome c, apoptosis-inducing factors (AIFs), and mitochondrial DNA, was restored to the almost normal levels by IL-1Ra pretreatment (Fig. 2I). Taken together, these results imply that IL-1β signaling plays a critical role in leptin-induced apoptosis of hepatocytes.
We further elucidated the detailed molecular mechanisms by which IL-1β signaling mediates apoptosis and cell cycle arrest. Given that AKT pathway plays a critical role in the survival of hepatocytes (Jing
In ensuing experiments to verify the functional role of AKT inactivation in leptin-induced hepatocytes death, leptin-stimulated annexin V/7-AAD binding was markedly restored by treatment with a pharmacological activator of AKT (SC79) (Fig. 3E). The formation of cleaved caspase-3 was also significantly suppressed by treatment with SC79 (Fig. 3F), indicating that AKT inactivation is implicated in leptin-induced apoptosis of hepatocyte. We further found that treatment with AKT activator reverted the changes in cell populations in each cell cycle phase (Fig. 3G) and suppression of p16 induction (Fig. 3H), implying that AKT inactivation also plays an important role in leptin-induced cell cycle arrest in hepatocytes.
Upon further exploration, we found that leptin treatment activated GSK3β, which is known as modulator of apoptosis in hepatocytes (Chen
p38MAPK, a typical stress-activated serine/threonine kinase, is considered a critical signaling molecule that modulates apoptosis and cell cycle arrest in hepatocytes (Awad
IL-1β-dependent signaling is conveyed by binding with its functional receptor (IL-1R1). To validate the role of IL-1β signaling in leptin-modulation of liver physiology
Leptin, a hormone primarily produced by the adipose tissue, was originally reported to play a crucial role in regulating energy balance. Recent studies have demonstrated that its receptors are present throughout the body and modulate a variety of physiological responses, apart from its classical metabolic actions. In particular, there is a growing appreciation that high plasma levels of leptin, termed hyperleptinemia, is closely associated with the development of liver diseases at multiple stages (Chitturi
Abnormal production of pro-inflammatory cytokines is closely associated with the initiation, development, and progression of various types of liver diseases. In most cases, pro-inflammatory cytokines in the liver originate from immune cells, such as Kupffer cells and other infiltrating immune cells (Luan
We have previously reported that leptin induces pyroptotic cell death in hepatocyte (Baral and Park, 2021). In this study, we also found that leptin induces apoptosis in cultured hepatocytes (Fig. 2). The results observed in the present study and previous reports imply that different types of cell death occur in hepatocytes upon exposure to leptin. Although apoptosis and pyroptosis are distinct types of programmed cell death, they are interconnected in certain conditions. Interestingly, inflammasomes activation has been shown to initially lead to pyroptotic cell death, accompanied by a release of inflammatory cytokines and pro-inflammatory cell death, followed by the apoptotic pathway (Wallace
AKT signaling is well known to play a critical role in the survival of liver cells. Leptin has been widely shown to phosphorylate AKT and activate its downstream signaling. For instance, leptin induces phosphorylation of AKT in cancer cells, which results in activation of various downstream signaling, leading to cancer cell growth (Saxena
In addition to the potent pro-inflammatory properties, IL-1β signaling has been shown to induce cell cycle arrest and apoptosis in various types of cells (Guadagno
In conclusion, data presented in this study demonstrated that IL-1β maturation and secretion upon NLRP3 inflammasomes activation critically contributes to leptin-induced cell cycle arrest and apoptosis in hepatocytes. Furthermore, we have elucidated that cytotoxic effects by IL-1β signaling is mediated via differential regulation of AKT and p38MAPK (Fig. 7). Given that leptin signaling is implicated in various types of hepatic injury, in addition to direct hepatotoxic effect, IL-1β would be a potential target for the treatment of liver pathologies associated with leptin.
This work was supported by the Yeungnam University research grant in 2022. The authors thank the Core Research Support Center for Natural Products and Medical Materials (CRCNM) for the technical support regarding the confocal microscopic analysis.
The authors declare that there are no competing interests.
Park PH; designed the study, analyzed the data, edit/wrote the manuscript. Baral A; performed the experiments, analyzed the data, wrote the manuscript.