Abstract
Hepatocellular carcinoma (HCC) is a disease with high morbidity, high mortality, and low cure rate. Hyaluronic acid (HA) is widely adopted in tissue engineering and drug delivery. 5-(4-Hydroxyphenyl)-3H-1, 2-dithiol-3-thione (ADT-OH) is one of commonly used H2S donors. In our previous study, HA-ADT was designed and synthesized via coupling of HA and ADT-OH. In this study, compared with sodium hydrosulfide (NaHS, a fast H2S-releasing donor) and morpholin-4-ium (4-methoxyphenyl)-morpholin-4-ylsulfanylidenesulfido-λ5-phosphane (GYY4137, a slow H2S-releasing donor), HA-ADT showed stronger inhibitory effect on the proliferation, migration, invasion, and cell cycle of human HCC cells. HA-ADT promoted apoptosis by suppressing the expressions of phospho (p)-protein kinase B (PKB/AKT), p‐glycogen synthase kinase‐3β (GSK-3β), p‐β‐catenin, and also inhibited autophagy via the downregulation of the protein levels of p‐Smad2, p‐Smad3, and transforming growth factor‐β (TGF‐β) in human HCC cells. Moreover, HA-ADT inhibited HCC xenograft tumor growth more effectively than both NaHS and GYY4137. Therefore, HA-ADT can suppress the growth of HCC cells by blocking the AKT/GSK-3β/β-catenin and TGF‐β/Smad2/3 signaling pathways. HA-ADT and its derivatives may be developed as promising antitumor drugs.
1. Introduction
Liver cancer is one of the leading causes of cancer death worldwide [1–4]. It is a heterogeneous, invasive, and drug-resistant malignant disease with poor prognosis [5–7], which is also the most common malignant tumor in the digestive system, with high mortality and low survival rate [3, 8]. Infection with hepatitis viruses and dietary exposure to aflatoxin are the main causes of liver cancer [9]. Hepatocellular carcinoma (HCC) is a highly invasive and most common liver cancer [10–12]. Many factors can contribute to the development of liver cancer, including hepatitis B virus, hepatitis C virus, cirrhosis, alcoholism, obesity, poor diet and inactivity, and mycotoxins [13–18]. Recent studies have shown undifferentiated liver cancer stem cells to be the main cause for the occurrence, metastasis, recurrence, and chemotherapy resistance of liver cancer [11, 19]. Liver cancer patients often die due to difficulties in early diagnosis, missed opportunity for surgical resection, and delayed treatment [3, 6]. Further research is urgent to advance the prevention, diagnosis, and treatment of liver cancer [13, 20].
Hydrogen sulfide (H2S) is a water-soluble and colorless gas with the rotten egg odor [21, 22]. The physiological function of H2S has been firstly demonstrated in the mammalian brain [23]. H2S has been identified as a gaseous signaling molecule, and together with carbon monoxide (CO) and nitric oxide (NO), forms a bioactive gas transmitter group [24–28]. H2S plays an important role in signal transduction in a variety of physiological and pathological processes [29–35]. Cystathionine γ-lyase (CSE), cystathionine β-synthetase (CBS), and 3-mercaptopyruvate sulfurtransferase (3-MST) are three main enzymes involved in endogenous H2S biosynthesis under physiological conditions [21, 32, 36, 37]. H2S breakdown is accomplished by a mitochondrial pathway that couples H2S oxidation into sulfate and thiosulfate to adenosine triphosphate synthesis [32]. In addition to the absorption of H2S through diffusion, a small amount of endogenous H2S can also be produced by the dietary L-homocysteine through the sulfur transfer pathway. This process is mainly completed by two enzymes, CBS and CSE, which depend on pyridoxal-5′-phosphate. CBS and CSE use cystathionine to convert homocysteine into cysteine, and H2S is a byproduct. In addition, 3-MST can also generate H2S. It uses mercaptopyruvate to form persulphide intermediately through α-ketoglutarate and cysteine transaminase, and then release H2S and pyruvate through reduction reaction [26, 29, 36, 38].
Hyaluronic acid (HA) is a glycosaminoglycan widely existing in human body. HA, a natural polysaccharide, is composed of N-acetylglucosamine and glucuronic acid, which are alternately linked by β-1, 3 and β-1, 4 glycoside bonds [39, 40]. Because HA and its derivatives have the characteristics of high viscoelasticity, plasticity, nonimmunogenicity, good biocompatibility, degradation, and binding with specific receptors on the cell surface, they are often used as slow-release carriers of drugs or as active target ligands of modified nano carriers to achieve the thickening, slow-release, transdermal absorption of drugs and improve the targeting and bioavailability of drugs [39–41]. HA and drug conjugates have been shown to have the dual advantages of tumor site aggregation and receptor-mediated endocytosis [40]. 5-(4-hydroxyphenyl)-3H-1, 2-dithiol-3-thione (ADT-OH) is the most widely used agent in the synthesis of slow H2S-releasing donors. ADT is a methyl derivative of ADT-OH, which can be metabolized by mitochondrial enzymes to produce H2S [42, 43].
A number of studies have shown that the protein kinase B (PKB/AKT)/glycogen synthase kinase‐3β (GSK-3β)/β-catenin pathway plays an important role in the progression of liver cancer [44–46]. Furthermore, the transforming growth factor‐β (TGF‐β)/Smad2/3 pathway is involved in the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of liver cancer [47–49]. Diallyl trisulfide, an H2S donor, regulates cell invasion and apoptosis via the phosphatidylinositol 3-kinase/AKT/GSK-3β signaling pathway in human osteosarcoma U2OS cells [50]. Another study indicates that H2S can attenuate paraquat-induced EMT of human alveolar epithelial cells by regulating the TGF-β1/Smad2/3 pathway [51]. Our previous study has demonstrated that H2S plays a double-edged sword role in human HCC cells, suggesting that novel H2S donors can be designed and applied for the treatment of cancer [52].
In this study, a new coupling compound HA-ADT was designed and synthesized as previous described [53]. HA-ADT could generate more H2S than both sodium hydrosulfide (NaHS, a fast H2S-releasing donor) and morpholin-4-ium (4-methoxyphenyl)-morpholin-4-ylsulfanylidenesulfido-λ5-phosphane (GYY4137, a slow H2S-releasing donor) [53]. The roles of HA-ADT in the proliferation, migration, and invasion of human HCC cells were investigated. Then we conducted in vivo experiments to further determine the effect of HA-ADT on the growth of human HCC xenografts.
2. Materials and Methods
2.1. Cell Culture
Human HCC cell lines SMMC-7721 and Huh-7 were obtained from Kebai Biological Technology Co., Ltd. (Nanjing, Jiangsu, China). SMMC-7721 cells were grown in RPMI1640 medium supplemented with 10% fetal bovine serum (FBS), penicillin (100 U/ml), and streptomycin (100 μg/mL). Huh-7 cells were grown in DMEM supplemented with 10% FBS, penicillin (100 U/ml), and streptomycin (100 μg/mL). Cells were cultured in an incubator at 37°C with 95% air and 5% CO2. Cells were treated with NaHS (200 μM), GYY4137 (200 μM), and HA-ADT (200 μM) (provided by Prof. Shao-Feng Duan), respectively [53]. The control group was treated with phosphate-buffered saline (PBS). After treatment for 24 h, the cells were adopted for following experiments.
2.2. Cell Growth Assay
The Cell-Light 5-ethynyl-2-deoxyuridine (EdU) Apollo 567 Kits (RiboBio, Guangzhou, China) were used to detect cell proliferation. Cell proliferation rate = (EdU-positive cells)/(total cells) × 100% [54]. The 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) (Sigma, St. Louis, MO, USA) and CCK-8 detection kits (Beyotime, Shanghai, China) were adopted to determine cell viability [55–57].
2.3. Colony Formation Assay
The cells (1 × 103 per well) were cultivated in a culture medium for 2 weeks at 37°C. After washing with PBS, the colonies were fixed with methanol. Crystal violet was then added and incubated at room temperature for 30 min. The plates were washed, air-dried, and scanned. Then, the colony number was counted.
2.4. Wound Healing Assay
The cultured monolayer-confluent cells were scratched. The migration distance was photographed under an inverted microscope (Olympus CKX41, Tokyo, Japan). The cell migration rate (MR) was calculated as MR (%) = [(A − B)/A] × 100, where A and B are the widths at 0 h and 24 h, respectively [58].
2.5. Migration and Invasion Assays
The cells (1 × 105) were seeded into the matrigel coated/uncoated upper chamber. The medium supplemented with 20% FBS was added into the lower chamber. After 24 h of treatment, the remaining cells on the upper side were scrubbed off, and the cells on the bottom side were fixed with 4% paraformaldehyde, and then stained with 0.1% crystal violet for 20 min. The cells were counted using an Axioskop 2 plus microscope (Zeiss, Thornwood, NY, USA).
2.6. Flow Cytometry Analysis
1 × 106 cells were trypsinized and then fixed in ice-cold 75% ethanol overnight. After washing with PBS, the cells were incubated in propidium iodide (PI)/RNase A mixture at room temperature for 30 min. The cell cycle distribution was analyzed with a FACSVerse flow cytometer (CytoFLEX S, Beckmann, CA, USA). The apoptotic level was detected using the Annexin V and PI apoptosis kits (UE, Suzhou, Jiangsu, China) and analyzed using a FACSVerse flow cytometer.
2.7. TdT-Mediated dUTP-Biotin Nick End Labeling (TUNEL) Assay
TUNEL assay was carried out using the in situ cell death detection kits (Beyotime). The cells were examined with a fluorescence microscope (Nikon Eclipse Ti, Melville, NY, USA). The percentage of TUNEL positive cells was further quantified.
2.8. Western Blotting
Total protein was extracted from human HCC cells. Western blotting was used to determine the expression levels of relevant proteins. The primary antibodies include anti-Cyclin E1, anti-Cyclin D1, anti-cyclin-dependent kinase (CDK) 2, anti-CDK4, anti-p27, anti-p21, anti-AKT, anti-phospho (p)-AKT (Ser473), anti-glycogen synthase kinase-3 beta (Gsk‐3β), anti-p-Gsk-3β (Ser9), anti-β-catenin, anti-p-β-catenin (Ser552), anti-beclin-1, anti-p62, anti-LC3A/B, anti-Smad2, anti-p-Smad2 (Ser465/467), anti-Smad3, anti-p-Smad3 (Ser423/425), and anti-transforming growth factor-beta (TGF‐β) antibodies, as well as the horseradish peroxidase-conjugated secondary antibody obtained from Cell Signaling Technology (CST, Danvers, MA, USA). Anti-B-cell lymphoma-2 (Bcl-2), anti-B-cell lymphoma-extra large (Bcl-xl), anti-Bcl-2-associated X protein (Bax), anti-Bcl-xl/Bcl-2-associated death promoter (Bad), anti-cleaved caspase (cas)-3, anti-cleaved cas-9, anti-cleaved poly adenosine diphosphate‐ribose polymerase (PARP), and anti-β-actin antibodies were obtained from ProteinTech (Chicago, IL, USA). The protein bands were detected with the enhanced chemiluminescence system (Thermo, Rockford, IL, USA) and semiquantified by ImageJ software.
2.9. Animal Study
Animal experiments were approved by the Committee of Medical Ethics and Welfare for Experimental Animals of Henan University School of Medicine (HUSOM-2017-218). Animal study was carried out as previously described [53]. BALB/c nude mice (male, 4-week-old) were obtained from Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). 5 × 106 SMMC-7721/Huh-7 cells in PBS (200 μL) were injected subcutaneously into the right flank of each mouse. The mice were divided randomly into 4 groups (n = 6 per group). NaHS (200 μM), GYY4137 (200 μM), HA-ADT (200 μM), and PBS were administered subcutaneously once-a-day for 21 days. During the animal experiment, the mice were daily weighed. The tumor volume was calculated as follows: Volume (V) = 1/2 × W2 × L, where L and W are the longest and widest dimension, respectively [59]. The tumor volume doubling time (TVDT) was calculated as follows: TVDT = (T − T0) × log2/log (V2/V1), where V2 and V1 are the tumor volume at two measurement times and (T − T0) is the time interval [60, 61]. At the end of the experiment, all mice were sacrificed. Then the tumors were excised, weighted, and photographed. Inhibition rate (IR) = [(A − B)/A] × 100%, where A and B are the average tumor weight of control group and the treatment group, respectively [53].
2.10. Hematoxylin and Eosin (HE) Staining
Tumor tissues were fixed in 10% neutral-buffered formalin, embedded in paraffin, sectioned at 5 μm thickness, and then stained with HE. The tissues were observed using a Zeiss Axioskop 2 plus microscope.
2.11. Immunohistochemistry (IHC)
Microvessel density (MVD) has been widely used as an index for the angiogenic activity [62]. The proliferation index (PI) was calculated as the percentage of Ki67-positive cells to total cells [63]. Apoptotic index was determined as cleaved cas-3 -positive cells to total cells [64]. Tumor tissues were stained with anti-CD31 (CST), anti-Ki67 (CST), anti-p21, anti-cleaved cas-3, and anti-beclin-1 antibodies, respectively. The proliferation index (PI), apoptosis index, MVD, p21-positive cells, and autophagy index were counted according to the ratio of positive cells to total cells.
2.12. Statistical Analysis
All results are expressed as the mean ± standard error of the mean (SEM). Differences among groups were determined using one-way analysis of variance with SPSS 19.0 software, followed by Tukey's test. A P value of less than 0.05 was considered statistically significant.
3. Results
3.1. HA-ADT Inhibits the Growth, Migration, and Invasion of Human HCC Cells
Compared with the control, NaHS, and GYY4137 group, HA-ADT significantly suppressed the viability and proliferation of Huh-7 and SMMC-7721 cells (Figures 1(a)–1(e)). HA-ADT also more effectively reduced the colony formation of SMMC-7721 and Huh-7 cells (Figures 1(f) and 1(g)). In addition, HA-ADT showed more inhibitory effects on the migration and invasion of Huh-7 and SMMC-7721 cells than the control, NaHS, and GYY4137 group (Figure 2). In summary, the results show that HA-ADT could inhibit the growth, migration, and invasion of HCC cells.
Figure 1.
Effects of HA-ADT on proliferative ability and cell viability of human HCC cells. (a) EdU assay was adopted to determine DNA replication activity (original magnification, 100x). (b) Cell proliferation rate was calculated. (c) Phase contrast microscopy of Huh-7 and SMMC-7721 cells (original magnification, 100x). (d, e) Cell viability was measured by MTT and CCK8 assays. Cell viability in the control group was considered to be 100%. (f) The cells were treated for 2 weeks and then the clonogenic ability was detected. (g) The number of colonies was calculated. All data are shown as the mean ± SEM of three independent experiments; ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
Figure 2.
Effects of HA-ADT on the migrative and invasive capacities of human HCC cells. (a) Wound-healing assay was performed to determine cell migration (original magnification, 100x). (b) The migration rate was determined. (c, d) Transwell assay was carried out to evaluate the migrative and invasive abilities of human HCC cells (original magnification, 200x). (e, f) The numbers of the migrative and invasive cells were determined. All data are shown as the mean ± SEM of three independent experiments; ∗P < 0.05, ∗∗P < 0.01 vs. control group; △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
3.2. HA-ADT Blocks Cell Cycle of Human HCC Cells
As shown in Figures 3(a) and 3(b), HA-ADT significantly upregulated the percentage of cells in S phase but downregulated the percentage of cells in G2 phase, suggesting that HA-ADT blocked the cell cycle in S phase. Western blot was further conducted to detect the protein levels of cyclin D1/E1, CDK2/4, p21, and p27. The results indicated that HA-ADT decreased the levels of cyclin D1/E1 and CDK2/4, but increased the levels of both p21 and p27 (Figures 3(c) and 3(d)). These results indicate that HA-ADT can block the cell cycle of human HCC cells at S phase by regulating the expression levels of cell cycle-related proteins.
Figure 3.
Effects of HA-ADT on the cell cycle of human HCC cells. (a) Cell cycle distribution was detected by flow cytometry. (b) The results of cell cycle distribution were analyzed. (c) The expression levels of cyclin D1/E1, CDK2/4, p21, and p27 were detected. β-actin was adopted as the internal control. (d) The band density was analyzed. All data are shown as the mean ± SEM of three independent experiments; ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
3.3. HA-ADT Promotes Apoptosis by Suppressing the AKT/GSK-3β/β-Catenin Signaling Pathway in Human HCC Cells
Apoptosis was detected by TUNEL and flow cytometry assays. As shown in Figures 4(a)–4(d), the apoptotic level in the HA-ADT group was significantly increased when compared with the control, NaHS, and GYY4137 group. The ratios of Bax/Bcl-2 and Bad/Bcl-xl are important indicators of apoptosis. Increased Bax/Bcl-2 and Bad/Bcl-xl ratios have been found in mitochondria-mediated apoptosis [65, 66]. As shown in Figure S1, Bax/Bcl-2 and Bad/Bcl-xl ratios in the HA-ADT group were obviously increased. In addition, the expression levels of cleaved cas-3, 9, and cleaved PARP exhibited similar trends. The AKT/GSK-3β/β-catenin cascade is a key pathway involved in the survival, growth, and metabolic stability of tumor cells [67, 68]. As shown in Figures 4(e) and 4(f), the phosphorylation levels of AKT, GSK-3β, and β-catenin were downregulated in the HA-ADT group. The data suggest that HA-ADT induces apoptosis via suppressing the AKT/GSK-3β/β-catenin pathway in human HCC cells.
Figure 4.
Effects of HA-ADT on the apoptotic level and AKT/GSK-3β/β-catenin pathway in human HCC cells. (a) TUNEL staining was used to detect the apoptotic level (original magnification, 100x). (b) The apoptotic index was counted as the ratio of TUNEL positive cells to total cells. (c) Flow cytometry assay was adopted to detect apoptosis. (d) Cell apoptosis distribution was analyzed. (e) Western blotting was used to determine the protein levels of AKT, p-AKT, GSK-3β, p-GSK-3β, β-catenin, and p-β-catenin. β-actin was adopted as the internal control. (f) The density was analyzed. All data are shown as the mean ± SEM of three independent experiments; ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
3.4. HA-ADT Decreases Autophagy of Human HCC Cells by Inhibiting the TGF-β/Smad2/3 Signaling Pathway
The role of autophagy in the development of cancer is extremely complex [69, 70]. Autophagy is a conservative catabolic process, which plays a dual role in regulating cell growth [71, 72]. Autophagy is an important mechanism by which cellular material is transfered to lysosome for degradation, thus providing energy and allowing the transformation of cellular components [70, 73, 74]. Beclin-1, LC3, and p62 are considered as specific autophagic markers [74]. As shown in Figures 5(a) and 5(b), the expressions of LC3 and beclin-1 in HA-ADT group were lower than those in the control, NaHS, and GYY4137 group, while the expression level of p62 exhibited the opposite trend. TGF-β plays an important role in cell homeostasis, fibrosis, angiogenesis, carcinogenesis, and differentiation. TGF-β can reduce apoptosis by inducing autophagy [75]. It has been shown that Smad2/3 is also involved in autophagy [76]. As shown in Figures 5(c) and 5(d), the expression levels of p-Smad2/3 and TGF-β in the HA-ADT group were lower than those in the control, NaHS, and GYY4137 group. These results indicate that HA-ADT can inhibit autophagy in human HCC cells via the TGF-β/Smad2/3 pathway.
Figure 5.
Effects of HA-ADT on the autophagic level and TGF-β/Smad2/3 signaling pathway in human HCC cells. (a) The protein levels of beclin-1, LC3A/B, and p62 were detected by western blotting. β-actin was adopted as the internal control. (b) The band density was analyzed. (c) The protein levels of p-Smad2, Smad2, p-Smad3, Smad3, and TGF-β were detected by western blotting. β-actin was adopted as the internal control. (d) The band density was analyzed. All data are shown as the mean ± SEM of three independent experiments; ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; #P < 0.05, ##P < 0.01 vs. GYY4137 group.
3.5. HA-ADT Suppresses the Growth of Human HCC Xenografted Tumors
SMMC-7721 and Huh-7 HCC cells have been successfully used to establish the subcutaneous xenograft models [77, 78]. Compared with the control, NaHS, and GYY4137 group, HA-ADT dramatically suppressed the growth of xenografted tumors (Figures 6(a)–6(e)). In addition, there was no significant difference in body weight between each group (Figures 6(f) and 6(g)). The expression levels of CD31, Ki67, p21, cleaved cas-3, and beclin-1 were further detected by IHC. As shown in Figure 7, the expression levels of CD31, Ki67, and beclin-1 in HA-ADT group were decreased, while p21 and cleaved cas-3 levels were increased in the HA-ADT group. These data demonstrate that HA-ADT can effectively suppress human HCC xenograft tumor growth by promoting apoptosis and reducing autophagy.
Figure 6.
Effects of HA-ADT on human HCC xenograft tumor growth. (a) Representative xenograft tumors of each group were shown. (b, c) The tumor volume and TVDT were determined. (d, e) The tumor weight and tumor inhibitory rate were measured. (f, g) The body weight change curve and the body weights of mice (day 0 and day 21) were calculated. All data are presented as the mean ± SEM (n = 6); ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
Figure 7.
Effects of HA-ADT on MVD, PI, cell cycle, apoptosis, and autophagy of human HCC xenograft tumors. (a–f) Representative photographs of HE staining, and IHC staining with anti-CD31, anti-Ki67, anti-p21, anti-cleaved cas-3, and anti-beclin-1 antibodies in human HCC xenograft tumors (original magnification, 400x). (g) The MVD, proliferation index, p21 positive cells, apoptotic index, and autophagic index were determined. All data are presented as the mean ± SEM (n = 6); ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
4. Discussion
At present, H2S is considered the third gaseous transmitter after CO and NO. H2S is involved in many physiological and pathological processes in the human body [21–23, 79]. HA and its derivatives have the characteristics of plasticity, nonimmunogenicity, and good biocompatibility, which are widely adopted in the biomedical field, such as tissue engineering and drug delivery [39–41]. ADT is a methyl derivative of ADT-OH, which is a common H2S donor [42, 79]. In the present study, HA-ADT was synthesized by chemical reaction as previously described [53].
Liver cancer is one of the leading causes of cancer death in the world [1–4]. Liver cancer is difficult to diagnose in the early stage, which will result in the death of patients. In addition, there are few effective treatments for patients with advanced liver cancer [3, 6, 13]. Thus, it is urgent to explore novel drugs to prevent and treat liver cancer. It has been revealed that 25–100 μM·NaHS promotes the growth of HCC cells and blood vessel formation, while 800–1000 μM·NaHS can inhibit angiogenesis and HCC growth [52]. Another study suggests that GYY4137 exhibits potent anti-HCC activity by blocking the signal transducer and activator of transcription 3 pathway [80]. In this study, we examined the roles of HA-ADT in the growth, migration, invasion, and cell cycle of human HCC cells. The results showed that HA-ADT was more effective than both NaHS and GYY4137 in inhibiting the survival, proliferation, migration, invasion, and cell cycle progression of human HCC cells. These results suggest that HA-ADT plays an effective role in inhibiting the development and progression of human HCC cells.
Apoptosis, a form of programmed cell death, is evolutionarily conserved and plays a key role in the homeostasis and development of mammalian tissues [81]. Apoptotic pathways can be divided into two categories: mitochondria-mediated intrinsic pathway and death receptor-mediated extrinsic pathway [82]. Bcl-2 family proteins are pivotal members in the process of apoptosis [53]. Caspases play key roles in apoptotic signaling pathways [83]. Caspases can be activated by many apoptotic stimuli and PARP is cleaved by cleaved caspase-3, resulting in the occurrence of apoptosis [53, 84]. It has been shown that NaHS effectively decreases the growth of C6 glioma cells by inducing caspase-dependent apoptosis [85]. Furthermore, GYY4137 could induce apoptosis in HCC cells by increasing the levels of cleaved cas-9, cas-3 and PARP cleavage [80]. Similarly, our results indicated that HA-ADT can promote apoptosis in HCC cells by up-regulating the levels of cleaved cas-3, 9, and cleaved PARP, indicating the activation of mitochondrial apoptosis. The AKT/GSK-3β/β-catenin pathway is involved in a number of hallmarks of cancer, such as tumor grade and lympho-node metastasis [67, 68, 86]. It has been reported that the AKT pathway is involved in HCC growth and metastasis [68]. In addition, GSK-3β plays a key role in the phosphorylation/degradation of β-catenin in the AKT/GSK-3β/β-catenin pathway [68]. The results suggested that HA-ADT could reduce the expressions of p-AKT, p-GSK-3β, and p-β-catenin in human liver cancer cells. The current research suggests that HA-ADT can inhibit the growth of human HCC cells by inducing apoptosis via inhibition of the AKT/GSK-3β/β-catenin signaling pathway.
Autophagy could be neutral, tumor-promoting, or tumor-suppressive in different contexts in cancer cells [87]. A recent study has shown that diallyl trisulfide, a characterized H2S donor, inhibits the proliferation of urothelial carcinoma cells by promoting apoptosis and inducing autophagy [88]. Our previous study has demonstrated that HA-ADT could suppress the progression of esophageal squamous cell carcinoma via apoptosis promotion and autophagy inhibition [89]. Similarly, in the present study, our data showed that HA-ADT significantly reduced the autophagic level when compared to the control, NaHS, and GYY4137 group. It has been reported that the TGF-β pathway can activate autophagy in many human cancer cells, suggesting that induction of autophagy is a novel biological function of TGF-β [90]. Furthermore, as canonical effectors of TGF-β signaling, Smad2/3 are involved in the process of autophagy [91]. Moreover, another study suggests that specificity protein 1-mediated serine/threonine kinase 39 upregulation promotes the proliferation, migration, and invasion of HCC cells by activating the TGF-β1/Smad2/3 pathway [47]. Our data indicated that HA-ADT decreased the expressions of p-Smad2/3 and TGF-β compared with the control, NaHS, and GYY4137 group. These findings indicate that HA-ADT can inhibit autophagy in human HCC cells through the TGF-β/Smad2/3 signaling pathway.
Recent studies suggest that SMMC-7721 and Huh-7 cells have been widely adopted to establish the xenograft tumor models [77, 78]. Therefore, we studied the role of HA-ADT in the growth of HCC xenograft tumor. We observed that HA-ADT exerted more inhibitory effects on HCC xenograft tumor growth than the control, NaHS, and GYY4137 group. Furthermore, there was no obvious change in the body weight in each group. Similar to the in vitro findings, our data suggested that the expressions of Ki67, CD31, and beclin-1 were decreased in HA-ADT group. The levels of p21 and cleaved cas-3 in the HA-ADT group were significantly higher than those in the control, NaHS, and GYY4137 group. The data together indicate that HA-ADT can effectively inhibit the growth of human HCC xenograft tumors.
In sum, HA-ADT can suppress the proliferation, migration and invasion of human HCC cells via inhibition of the AKT/GSK-3β/β-catenin and TGF-β/Smad2/3 signaling pathways. HA-ADT might be considered as a promising anticancer candidate for the treatment of HCC.
Acknowledgments
The study was supported by grants from the National Natural Science Foundation of China (no. 81802718), the Natural Science Foundation of Henan Province, China (no. 162300410044), the Foundation of Science & Technology Department of Henan Province, China (no. 202102310480), the Training Program for Young Backbone Teachers of Institutions of Higher Learning in Henan Province, China (no. 2020GGJS038), and the Science Foundation for Young Talents of Henan University College of Medicine, China (no. 2019013).
Contributor Information
Dong-Dong Wu, Email: ddwubiomed2010@163.com.
Xiao-Ju Zhang, Email: 15837101166@163.com.
Juan Cen, Email: cenjuan@vip.henu.edu.cn.
Data Availability
All data generated or analyzed in this study are included in this article.
Conflicts of Interest
The authors declare that they have no conflicts of interest.
Authors' Contributions
D.D.W., X.J.Z., and J.C. participated in the conception and design of the experiments. S.F.D., M.M.Z., Q. D., B.Y., W.L., X.Z., H.L.Y., S.H.Z., and N.H.K. carried out the experiments and analyzed the data. D.D.W. and S.F.D. prepared the manuscript. All authors read and approved the final manuscript. S.F.D and M.M.Z equally contributed to this study.
Supplementary Materials
Figure S1: Effects of HA-ADT on the expression levels of mitochondrial apoptosis-related proteins in human HCC cells. (a) Western blotting was used to detect the expressions of Bcl-2, Bax, Bcl-xl, Bad, cleaved cas-3, 9, and cleaved PARP in SMMC-7721 and Huh-7 cells. β-actin was adopted as the internal control. (b) The band density was analyzed. The Bax/Bcl-2 and Bad/Bcl-xl ratios were calculated. All data are shown as the mean ± SEM of three independent experiments; ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
References
- 1.Ma X., Tan Y. T., Yang Y., et al. Pre-diagnostic urinary 15-F2t -isopros tane level and liver cancer risk: results from the Shanghai Women’s and Men’s Health Studies. International Journal of Cancer . 2018;143(8):1896–1903. doi: 10.1002/ijc.31591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Maucort-Boulch D., de Martel C., Franceschi S., Plummer M. Fraction and incidence of liver cancer attributable to hepatitis B and C viruses worldwide. International Journal of Cancer . 2018;142(12):2471–2477. doi: 10.1002/ijc.31280. [DOI] [PubMed] [Google Scholar]
- 3.Fu J., Wang H. Precision diagnosis and treatment of liver cancer in China. Cancer Letters . 2018;412:283–288. doi: 10.1016/j.canlet.2017.10.008. [DOI] [PubMed] [Google Scholar]
- 4.Cheng Z., Lei Z., Yang P., et al. Long non-coding RNA THOR promotes liver cancer stem cells expansion via β-catenin pathway. Gene . 2019;684:95–103. doi: 10.1016/j.gene.2018.10.051. [DOI] [PubMed] [Google Scholar]
- 5.Li L., Wang H. Heterogeneity of liver cancer and personalized therapy. Cancer Letters . 2016;379(2):191–197. doi: 10.1016/j.canlet.2015.07.018. [DOI] [PubMed] [Google Scholar]
- 6.Cheng Z., Li X., Ding J. Characteristics of liver cancer stem cells and clinical correlations. Cancer Letters . 2016;379(2):230–238. doi: 10.1016/j.canlet.2015.07.041. [DOI] [PubMed] [Google Scholar]
- 7.Gailhouste L., Liew L. C., Yasukawa K., et al. Differentiation therapy by epigenetic reconditioning exerts antitumor effects on liver cancer cells. Molecular Therapy . 2018;26(7):1840–1854. doi: 10.1016/j.ymthe.2018.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zhou Y., Li Y., Zhou T., Zheng J., Li S., Li H. B. Dietary natural products for prevention and treatment of liver cancer. Nutrients . 2016;8(3):p. 156. doi: 10.3390/nu8030156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kensler T. W., Qian G. S., Chen J. G., Groopman J. D. Translational strategies for cancer prevention in liver. Nature Reviews Cancer . 2003;3(5):321–329. doi: 10.1038/nrc1076. [DOI] [PubMed] [Google Scholar]
- 10.Marengo A., Rosso C., Bugianesi E. Liver cancer: connections with obesity, fatty liver, and cirrhosis. Annual Review of Medicine . 2016;67(1):103–117. doi: 10.1146/annurev-med-090514-013832. [DOI] [PubMed] [Google Scholar]
- 11.Lv H., Lv G., Han Q., Yang W., Wang H. Noncoding RNAs in liver cancer stem cells: the big impact of little things. Cancer Letters . 2018;418:51–63. doi: 10.1016/j.canlet.2018.01.001. [DOI] [PubMed] [Google Scholar]
- 12.Samson A., Bentham M. J., Scott K., et al. Oncolytic reovirus as a combined antiviral and anti-tumour agent for the treatment of liver cancer. Gut . 2018;67(3):562–573. doi: 10.1136/gutjnl-2016-312009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Michelotti G. A., Machado M. V., Diehl A. M. NAFLD, NASH and liver cancer. Nature Reviews Gastroenterology & Hepatology . 2013;10(11):656–665. doi: 10.1038/nrgastro.2013.183. [DOI] [PubMed] [Google Scholar]
- 14.Duan X. Y., Zhang L., Fan J. G., Qiao L. NAFLD leads to liver cancer: do we have sufficient evidence? Cancer Letters . 2014;345(2):230–234. doi: 10.1016/j.canlet.2013.07.033. [DOI] [PubMed] [Google Scholar]
- 15.Nikolaou K., Sarris M., Talianidis I. Molecular pathways: the complex roles of inflammation pathways in the development and treatment of liver cancer. Clinical Cancer Research . 2013;19(11):2810–2816. doi: 10.1158/1078-0432.ccr-12-1961. [DOI] [PubMed] [Google Scholar]
- 16.Pang Y., Kartsonaki C., Turnbull I., et al. Diabetes, plasma glucose, and incidence of fatty liver, cirrhosis, and liver cancer: a prospective study of 0.5 million people. Hepatology . 2018;68(4):1308–1318. doi: 10.1002/hep.30083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Liu X., Baecker A., Wu M., et al. Interaction between tobacco smoking and hepatitis B virus infection on the risk of liver cancer in a Chinese population. International Journal of Cancer . 2018;142(8):1560–1567. doi: 10.1002/ijc.31181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Nakagawa H., Fujita M., Fujimoto A. Genome sequencing analysis of liver cancer for precision medicine. Seminars in Cancer Biology . 2019;55:120–127. doi: 10.1016/j.semcancer.2018.03.004. [DOI] [PubMed] [Google Scholar]
- 19.Yang C., Cai W. C., Dong Z. T., et al. lncARSR promotes liver cancer stem cells expansion via STAT3 pathway. Gene . 2019;687:73–81. doi: 10.1016/j.gene.2018.10.087. [DOI] [PubMed] [Google Scholar]
- 20.Bruix J., Han K. H., Gores G., Llovet J. M., Mazzaferro V. Liver cancer: approaching a personalized care. Journal of Hepatology . 2015;62(1):S144–S156. doi: 10.1016/j.jhep.2015.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Suzuki K., Sagara M., Aoki C., Tanaka S., Aso Y. Clinical implication of plasma hydrogen sulfide levels in Japanese patients with type 2 diabetes. Internal Medicine . 2017;56(1):17–21. doi: 10.2169/internalmedicine.56.7403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Li M., Xu C., Shi J., et al. Fatty acids promote fatty liver disease via the dysregulation of 3-mercaptopyruvate sulfurtransferase/hydrogen sulfide pathway. Gut . 2018;67(12):2169–2180. doi: 10.1136/gutjnl-2017-313778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wang S., Liu X., Zhang M. Reduction of ammineruthenium(III) by sulfide enables in vivo electrochemical monitoring of free endogenous hydrogen sulfide. Analytical Chemistry . 2017;89(10):5382–5388. doi: 10.1021/acs.analchem.7b00069. [DOI] [PubMed] [Google Scholar]
- 24.Luo Z. L., Ren J. D., Huang Z., et al. The role of exogenous hydrogen sulfide in free fatty acids induced inflammation in macrophages. Cellular Physiology and Biochemistry . 2017;42(4):1635–1644. doi: 10.1159/000479405. [DOI] [PubMed] [Google Scholar]
- 25.Narne P., Pandey V., Phanithi P. B. Role of nitric oxide and hydrogen sulfide in ischemic stroke and the emergent epigenetic underpinnings. Molecular Neurobiology . 2019;56(3):1749–1769. doi: 10.1007/s12035-018-1141-6. [DOI] [PubMed] [Google Scholar]
- 26.Wetzel M. D., Wenke J. C. Mechanisms by which hydrogen sulfide attenuates muscle function following ischemia-reperfusion injury: effects on Akt signaling, mitochondrial function, and apoptosis. Journal of Translational Medicine . 2019;17(1):p. 33. doi: 10.1186/s12967-018-1753-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dulac M., Melet A., Galardon E. Reversible detection and quantification of hydrogen sulfide by fluorescence using the hemoglobin I from lucina pectinata. ACS Sensors . 2018;3(10):2138–2144. doi: 10.1021/acssensors.8b00701. [DOI] [PubMed] [Google Scholar]
- 28.Szijártó I. A., Markó L., Filipovic M. R., et al. Cystathionine γ-lyase-produced hydrogen sulfide controls endothelial NO bioavailability and blood pressure. Hypertension . 2018;71(6):1210–1217. doi: 10.1161/hypertensionaha.117.10562. [DOI] [PubMed] [Google Scholar]
- 29.Zhao Y., Henthorn H. A., Pluth M. D. Kinetic insights into hydrogen sulfide delivery from caged-carbonyl sulfide isomeric donor platforms. Journal of the American Chemical Society . 2017;139(45):16365–16376. doi: 10.1021/jacs.7b09527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Qiao Z., Zhang H., Wang K., Zhang Y. A highly sensitive and responsive fluorescent probe based on 6-azide-chroman dye for detection and imaging of hydrogen sulfide in cells. Talanta . 2019;195:850–856. doi: 10.1016/j.talanta.2018.12.014. [DOI] [PubMed] [Google Scholar]
- 31.Huang Z., He K., Song Z., et al. Alleviation of heavy metal and silver nanoparticle toxicity and enhancement of their removal by hydrogen sulfide in Phanerochaete chrysosporium. Chemosphere . 2019;224:554–561. doi: 10.1016/j.chemosphere.2019.02.190. [DOI] [PubMed] [Google Scholar]
- 32.Malagrinò F., Zuhra K., Mascolo L., et al. Hydrogen sulfide oxidation: adaptive changes in mitochondria of SW480 colorectal cancer cells upon exposure to hypoxia. Oxidative Medicine and Cellular Longevity . 2019;2019:11. doi: 10.1155/2019/8102936.8102936 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Barton M., Meyer M. R. HuR-ry up: how hydrogen sulfide protects against atherosclerosis. Circulation . 2019;139(1):115–118. doi: 10.1161/circulationaha.118.036854. [DOI] [PubMed] [Google Scholar]
- 34.Luo Y., Song Y., Zhu C., et al. Visualization of endogenous hydrogen sulfide in living cells based on Au nanorods@silica enhanced fluorescence. Analytica Chimica Acta . 2019;1053:81–88. doi: 10.1016/j.aca.2018.12.008. [DOI] [PubMed] [Google Scholar]
- 35.Youssef S., Zhang S., Ai H. W. A genetically encoded, ratiometric fluorescent biosensor for hydrogen sulfide. ACS Sensors . 2019;4(6):1626–1632. doi: 10.1021/acssensors.9b00400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hine C., Kim H. J., Zhu Y., et al. Hypothalamic-pituitary Axis regulates hydrogen sulfide production. Cell Metabolism . 2017;25(6):1320–1333.e5. doi: 10.1016/j.cmet.2017.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rodrigues C., Percival S. S. Immunomodulatory effects of glutathione, garlic derivatives, and hydrogen sulfide. Nutrients . 2019;11(2):p. 295. doi: 10.3390/nu11020295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Behera J., George A. K., Voor M. J., Tyagi S. C., Tyagi N. Hydrogen sulfide epigenetically mitigates bone loss through OPG/RANKL regulation during hyperhomocysteinemia in mice. Bone . 2018;114:90–108. doi: 10.1016/j.bone.2018.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Huang G., Huang H. Hyaluronic acid-based biopharmaceutical delivery and tumor-targeted drug delivery system. Journal of Controlled Release . 2018;278:122–126. doi: 10.1016/j.jconrel.2018.04.015. [DOI] [PubMed] [Google Scholar]
- 40.Huang G., Huang H. Application of hyaluronic acid as carriers in drug delivery. Drug Delivery . 2018;25(1):766–772. doi: 10.1080/10717544.2018.1450910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Singh A., Corvelli M., Unterman S. A., Wepasnick K. A., McDonnell P., Elisseeff J. H. Enhanced lubrication on tissue and biomaterial surfaces through peptide-mediated binding of hyaluronic acid. Nature Materials . 2014;13(10):988–995. doi: 10.1038/nmat4048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jia J., Xiao Y., Wang W., et al. Differential mechanisms underlying neuroprotection of hydrogen sulfide donors against oxidative stress. Neurochemistry International . 2013;62(8):1072–1078. doi: 10.1016/j.neuint.2013.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Montoya L. A., Pluth M. D. Organelle-Targeted H2S probes enable visualization of the subcellular distribution of H2S donors. Analytical Chemistry . 2016;88(11):5769–5774. doi: 10.1021/acs.analchem.6b00087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liao M., Du H., Wang B., Huang J., Huang D., Tong G. Anticancer effect of polyphyllin I in suppressing stem cell-like properties of hepatocellular carcinoma via the AKT/GSK-3β/β-catenin signaling pathway. Oxidative Medicine and Cellular Longevity . 2022;2022:23. doi: 10.1155/2022/4031008.4031008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhuang L., Wang X., Wang Z., et al. MicroRNA-23b functions as an oncogene and activates AKT/GSK3β/β-catenin signaling by targeting ST7L in hepatocellular carcinoma. Cell Death & Disease . 2017;8(5) doi: 10.1038/cddis.2017.216.e2804 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zheng W., Yao M., Wu M., Yang J., Yao D., Wang L. Secretory clusterin promotes hepatocellular carcinoma progression by facilitating cancer stem cell properties via AKT/GSK-3β/β-catenin axis. Journal of Translational Medicine . 2020;18(1):p. 81. doi: 10.1186/s12967-020-02262-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang J., Fan Z., Li J., Yang J., Liu X., Cheng J. Transcription factor specificity protein 1-mediated Serine/threonine kinase 39 upregulation promotes the proliferation, migration, invasion and epithelial-mesenchymal transition of hepatocellular carcinoma cells by activating the transforming growth factor-β1/Smad2/3 pathway. Bioengineered . 2021;12(1):3566–3577. doi: 10.1080/21655979.2021.1947939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhang M., Zheng S., Jing C., et al. S100A11 promotes TGF-β1-inducedepithelial-mesenchymal transition through SMAD2/3 signaling pathway in intrahepatic cholangiocarcinoma. Future Oncology . 2018;14(9):837–847. doi: 10.2217/fon-2017-0534. [DOI] [PubMed] [Google Scholar]
- 49.Shi J. Y., Ma L. J., Zhang J. W., et al. FOXP3 Is a HCC suppressor gene and Acts through regulating the TGF-β/Smad2/3 signaling pathway. BMC Cancer . 2017;17(1):p. 648. doi: 10.1186/s12885-017-3633-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.He P., Wang Z., Sheng B., et al. Diallyl trisulfide regulates cell apoptosis and invasion in human osteosarcoma U2OS cells through regulating PI3K/AKT/GSK3β signaling pathway. Histology & Histopathology . 2020;35(12):1511–1520. doi: 10.14670/HH-18-299. [DOI] [PubMed] [Google Scholar]
- 51.Bai Y. W., Ye M. J., Yang D. L., Yu M. P., Zhou C. F., Shen T. Hydrogen sulfide attenuates paraquat-inducedepithelial-mesenchymal transition of human alveolar epithelial cells through regulating transforming growth factor-β1/Smad2/3 signaling pathway. Journal of Applied Toxicology . 2019;39(3):432–440. doi: 10.1002/jat.3734. [DOI] [PubMed] [Google Scholar]
- 52.Wu D., Li M., Tian W., et al. Hydrogen sulfide acts as a double-edged sword in human hepatocellular carcinoma cells through EGFR/ERK/MMP-2 and PTEN/AKT signaling pathways. Scientific Reports . 2017;7(1):p. 5134. doi: 10.1038/s41598-017-05457-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Dong Q., Yang B., Han J. G., et al. A novel hydrogen sulfide-releasing donor, HA-ADT, suppresses the growth of human breast cancer cells through inhibiting the PI3K/AKT/mTOR and Ras/Raf/MEK/ERK signaling pathways. Cancer Letters . 2019;455:60–72. doi: 10.1016/j.canlet.2019.04.031. [DOI] [PubMed] [Google Scholar]
- 54.Zhang G. Y., Lu D., Duan S. F., et al. Hydrogen sulfide alleviates lipopolysaccharide-induced diaphragm dysfunction in rats by reducing apoptosis and inflammation through ROS/MAPK and TLR4/NF-κB signaling pathways. Oxidative Medicine and Cellular Longevity . 2018;2018:15. doi: 10.1155/2018/9647809.9647809 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Quarta A., Amorín M., Aldegunde M. J., et al. Novel synthesis of platinum complexes and their intracellular delivery to tumor cells by means of magnetic nanoparticles. Nanoscale . 2019;11(48):23482–23497. doi: 10.1039/c9nr07015j. [DOI] [PubMed] [Google Scholar]
- 56.Wu D., Zhong P., Wang J., Wang H. Exogenous hydrogen sulfide mitigates LPS + ATP-induced inflammation by inhibiting NLRP3 inflammasome activation and promoting autophagy in L02 cells. Molecular and Cellular Biochemistry . 2019;457(1-2):145–156. doi: 10.1007/s11010-019-03519-6. [DOI] [PubMed] [Google Scholar]
- 57.Zhang J., Si H., Li B., Zhou X., Zhang J. Myrislignan exhibits activities against toxoplasma gondii RH strain by triggering mitochondrial dysfunction. Frontiers in Microbiology . 2019;10:p. 2152. doi: 10.3389/fmicb.2019.02152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wu D. D., Gao Y. R., Li T., et al. PEST-containing nuclear protein mediates the proliferation, migration, and invasion of human neuroblastoma cells through MAPK and PI3K/AKT/mTOR signaling pathways. BMC Cancer . 2018;18(1):p. 499. doi: 10.1186/s12885-018-4391-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wang D. Y., Hong Y., Chen Y. G., et al. PEST-containing nuclear protein regulates cell proliferation, migration, and invasion in lung adenocarcinoma. Oncogenesis . 2019;8(3):p. 22. doi: 10.1038/s41389-019-0132-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Manesis E. K., Giannoulis G., Zoumboulis P., Vafiadou I., Hadziyannis S. J. Treatment of hepatocellular carcinoma with combined suppression and inhibition of sex hormones: a randomized, controlled trial. Hepatology . 1995;21(6):1535–1542. doi: 10.1016/0270-9139(95)90456-5. [DOI] [PubMed] [Google Scholar]
- 61.Wu D., Li J., Zhang Q., et al. Exogenous hydrogen sulfide regulates the growth of human thyroid carcinoma cells. Oxidative Medicine and Cellular Longevity . 2019;2019:18. doi: 10.1155/2019/6927298.6927298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Dong P., Fu H., Chen L., et al. PCNP promotes ovarian cancer progression by accelerating β-catenin nuclear accumulation and triggering EMT transition. Journal of Cellular and Molecular Medicine . 2020;24(14):8221–8235. doi: 10.1111/jcmm.15491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wu D. D., Liu S. Y., Gao Y. R., et al. Tumour necrosis factor-α-induced protein 8-like 2 is a novel regulator of proliferation, migration, and invasion in human rectal adenocarcinoma cells. Journal of Cellular and Molecular Medicine . 2019;23(3):1698–1713. doi: 10.1111/jcmm.14065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Huang J. S., Yang C. M., Wang J. S., et al. Caspase-3 expression in tumorigenesis and prognosis of buccal mucosa squamous cell carcinoma. Oncotarget . 2017;8(48):84237–84247. doi: 10.18632/oncotarget.20494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Green D. R., Llambi F. Cell death signaling. Cold Spring Harbor Perspectives in Biology . 2015;7(12) doi: 10.1101/cshperspect.a006080.a006080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Pitchakarn P., Suzuki S., Ogawa K., et al. Induction of G1 arrest and apoptosis in androgen-dependent human prostate cancer by Kuguacin J, a triterpenoid from Momordica charantia leaf. Cancer Letters . 2011;306(2):142–150. doi: 10.1016/j.canlet.2011.02.041. [DOI] [PubMed] [Google Scholar]
- 67.Wang C., Jin H., Wang N., et al. Gas6/Axl Axis contributes to chemoresistance and metastasis in breast cancer through akt/GSK-3β/β-catenin signaling. Theranostics . 2016;6(8):1205–1219. doi: 10.7150/thno.15083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Xu Q., Xu H. X., Li J. P., et al. Growth differentiation factor 15 induces growth and metastasis of human liver cancer stem-like cells via AKT/GSK-3β/β-catenin signaling. Oncotarget . 2017;8(10):16972–16987. doi: 10.18632/oncotarget.15216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pan H., Wang Y., Na K., et al. Autophagic flux disruption contributes to Ganoderma lucidum polysaccharide-induced apoptosis in human colorectal cancer cells via MAPK/ERK activation. Cell Death & Disease . 2019;10(6):p. 456. doi: 10.1038/s41419-019-1653-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Levy J. M. M., Towers C. G., Thorburn A. Targeting autophagy in cancer. Nature Reviews Cancer . 2017;17(9):528–542. doi: 10.1038/nrc.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Niu J., Yan T., Guo W., Wang W., Zhao Z. Insight into the role of autophagy in osteosarcoma and its therapeutic implication. Frontiers in Oncology . 2019;9:p. 1232. doi: 10.3389/fonc.2019.01232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wang L., Howell M. E. A., McPeak B., et al. LIMD1 is induced by and required for LMP1 signaling, and protects EBV-transformed cells from DNA damage-induced cell death. Oncotarget . 2017;9(5):6282–6297. doi: 10.18632/oncotarget.23676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mizushima N., Komatsu M. Autophagy: renovation of cells and tissues. Cell . 2011;147(4):728–741. doi: 10.1016/j.cell.2011.10.026. [DOI] [PubMed] [Google Scholar]
- 74.Tian L., Zhao Z., Xie L., Zhu J. MiR-361-5p suppresses chemoresistance of gastric cancer cells by targeting FOXM1 via the PI3K/Akt/mTOR pathway. Oncotarget . 2017;9(4):4886–4896. doi: 10.18632/oncotarget.23513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ding Y., Kim J. K., Kim S. I., et al. TGF-β1 protects against mesangial cell apoptosis via induction of autophagy. Journal of Biological Chemistry . 2010;285(48):37909–37919. doi: 10.1074/jbc.m109.093724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Pokharel S. M., Shil N. K., Bose S. Autophagy, TGF-β, and SMAD-2/3 signaling regulates interferon-β response in respiratory syncytial virus infected macrophages. Frontiers in Cellular and Infection Microbiology . 2016;6:p. 174. doi: 10.3389/fcimb.2016.00174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liao J., Jin H., Li S., et al. Apatinib potentiates irradiation effect via suppressing PI3K/AKT signaling pathway in hepatocellular carcinoma. Journal of Experimental & Clinical Cancer Research . 2019;38(1):p. 454. doi: 10.1186/s13046-019-1419-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhang X., Liu T., Li Z., et al. Hepatomas are exquisitely sensitive to pharmacologic ascorbate (P-AscH -) Theranostics . 2019;9(26):8109–8126. doi: 10.7150/thno.35378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Liu Y., Li J., Chen H., et al. Magnet-activatable nanoliposomes as intracellular bubble microreactors to enhance drug delivery efficacy and burst cancer cells. Nanoscale . 2019;11(40):18854–18865. doi: 10.1039/c9nr07021d. [DOI] [PubMed] [Google Scholar]
- 80.Lu S., Gao Y., Huang X., Wang X. GYY4137, a hydrogen sulfide (H2S) donor, shows potent anti-hepatocellular carcinoma activity through blocking the STAT3 pathway. International Journal of Oncology . 2014;44(4):1259–1267. doi: 10.3892/ijo.2014.2305. [DOI] [PubMed] [Google Scholar]
- 81.Cheng X., Ferrell J. E. Apoptosis propagates through the cytoplasm as trigger waves. Science . 2018;361(6402):607–612. doi: 10.1126/science.aah4065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhang P., Zhang Y., Liu K., et al. Ivermectin induces cell cycle arrest and apoptosis of HeLa cells via mitochondrial pathway. Cell Proliferation . 2019;52(2) doi: 10.1111/cpr.12543.e12543 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bai Y., Liu X., Qi X., et al. PDIA6 modulates apoptosis and autophagy of non-small cell lung cancer cells via the MAP4K1/JNK signaling pathway. EBioMedicine . 2019;42:311–325. doi: 10.1016/j.ebiom.2019.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hsu H. Y., Lin T. Y., Hu C. H., Shu D. T. F., Lu M. K. Fucoidan upregulates TLR4/CHOP-mediatedcaspase-3 and PARP activation to enhance cisplatin-induced cytotoxicity in human lung cancer cells. Cancer Letters . 2018;432:112–120. doi: 10.1016/j.canlet.2018.05.006. [DOI] [PubMed] [Google Scholar]
- 85.Zhao L., Wang Y., Yan Q., Lv W., Zhang Y., He S. Exogenous hydrogen sulfide exhibits anti-cancer effects though p38 MAPK signaling pathway in C6 glioma cells. Biological Chemistry . 2015;396(11):1247–1253. doi: 10.1515/hsz-2015-0148. [DOI] [PubMed] [Google Scholar]
- 86.Yu J., Wang X., Lu Q., et al. Extracellular 5’-nucleotidase (CD73) promotes human breast cancer cells growth through AKT/GSK-3β/β-catenin/cyclinD1 signaling pathway. International Journal of Cancer . 2018;142(5):959–967. doi: 10.1002/ijc.31112. [DOI] [PubMed] [Google Scholar]
- 87.Amaravadi R., Kimmelman A. C., White E. Recent insights into the function of autophagy in cancer. Genes & Development . 2016;30(17):1913–1930. doi: 10.1101/gad.287524.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Panza E., Bello I., Smimmo M., et al. Endogenous and exogenous hydrogen sulfide modulates urothelial bladder carcinoma development in human cell lines. Biomedicine & Pharmacotherapy . 2022;151 doi: 10.1016/j.biopha.2022.113137.113137 [DOI] [PubMed] [Google Scholar]
- 89.Duan S. F., Zhang M. M., Zhang X., et al. HA-ADT suppresses esophageal squamous cell carcinoma progression via apoptosis promotion and autophagy inhibition. Experimental Cell Research . 2022;420(1) doi: 10.1016/j.yexcr.2022.113341.113341 [DOI] [PubMed] [Google Scholar]
- 90.Kiyono K., Suzuki H. I., Matsuyama H., et al. Autophagy is activated by TGF-beta and potentiates TGF-beta-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Research . 2009;69(23):8844–8852. doi: 10.1158/0008-5472.can-08-4401. [DOI] [PubMed] [Google Scholar]
- 91.Pan C. C., Kumar S., Shah N., et al. Endoglin regulation of Smad2 function mediates Beclin1 expression and endothelial autophagy. Journal of Biological Chemistry . 2015;290(24):14884–14892. doi: 10.1074/jbc.m114.630178. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figure S1: Effects of HA-ADT on the expression levels of mitochondrial apoptosis-related proteins in human HCC cells. (a) Western blotting was used to detect the expressions of Bcl-2, Bax, Bcl-xl, Bad, cleaved cas-3, 9, and cleaved PARP in SMMC-7721 and Huh-7 cells. β-actin was adopted as the internal control. (b) The band density was analyzed. The Bax/Bcl-2 and Bad/Bcl-xl ratios were calculated. All data are shown as the mean ± SEM of three independent experiments; ∗P < 0.05, ∗∗P < 0.01 vs. control group; △P < 0.05, △△P < 0.01 vs. NaHS group; ##P < 0.01 vs. GYY4137 group.
Data Availability Statement
All data generated or analyzed in this study are included in this article.