scholarly journals UNDERSTANDING THE MECHANISM OF DRUG-RESISTANT AND TUMOR RECURRENCE IN LIVER CANCER

2018 ◽  
Vol 8 (5) ◽  
pp. 224-229
Author(s):  
Vasanthakumar Sekar ◽  
Muthu Dhandapani ◽  
Babu Balakrishnan ◽  
Kaleeswaran Balasubramanian ◽  
Sivalingam Azhagu Madhavan ◽  
...  

Chemo-resistant and tumor recurrence are the major hurdle to overcome the cancer patients. Especially in hepatocellular carcinoma (HCC) is notoriously refractory to chemotherapy because of its tendency to develop multi-drug resistance (MDR), through various mechanisms. Aim: The current research is focussed on understanding the mechanism involved in chemo-resistant and tumor recurrence in liver cancer. Methods: Human hepatocellular carcinoma cell line (Huh7) was used entire study. Huh7 cells were cultured with known chemotherapeutic drugs such as 5-FU, Paclitaxel and Cisplatin-based on their Cmax concentration, and then these drug-treated cells were examined for chemoresistant and tumor recurrence properties through flow cytometry analysis, spheroid formation assay, and morphological analysis. Results: In morphological analysis confirm these all the chemo drugs were shown more cytotoxic effete than control, even though there were few viable cells noticed in cisplatin treatment. In flow cytometry analysis cisplatin pre-treated cells were well expressed LCSC marker such as CD133 and stem cell transduction factors like Oct-4 & Nanog than control. In addition to this, all the CD133 expressed cells also expressed to EpCAM. In spheroid formation assay, cisplatin pre-treated cells shown well-defined spheroid than control. Conclusion: LCSC plays a major role in chemoresistant and tumor recurrence through PI3K/Akt/mTOR, wnt-β catenin signaling, NF-kB signaling. So, targeting LCSC through EpCAM targeted therapy along with chemotherapy might be the better option for enhanced prognosis. Keywords: LCSC, Chemoresistant, Tumor recurrence, Hepatocellular carcinoma.

2020 ◽  
Vol 9 ◽  
pp. 1896
Author(s):  
Maedeh Olya ◽  
Hamid Zaferani Arani ◽  
Amirhossein Shekarriz ◽  
Amirhossein Zabolian ◽  
Hadi Zare Marzouni ◽  
...  

Background: Hepatocellular carcinoma is the most common type of liver cancer which arises from the main cells in the liver. We address many studies investigating anti-cancer role of hypericin, however the proposing corresponding molecular pathway seems to be still a debate. Therefore, the present study aimed to evaluate the apoptotic effect of hypericin on the Huh7 as the liver cancer cell line and its relation with the gate keeper gene P53. Materials and Methods: In this study, the Huh7 cell line and fibroblast cells (as control group) were treated with different concentrations of hypericin for 24 and 48 hours. Detection of cell death was performed by MTT assay and flow cytometry. The expression of bax, bcl2 and p53 mRNAs was evaluated by Real-time PCR. Also, Immunocytochemistry (ICC) analysis was used for further evaluation of P53expression. Results: The results showed that hypericin has a dose-dependent cytotoxic effect on the Huh7 cell line, with no or marginal effect on fibroblastic cells. According to flow cytometry results, about 53%cells underwent apoptosis after exposure to LD50 of hypericin for 24 hours. Real-time PCR data demonstrated that the pro-apoptotic genes Bax and P53 expression level increased. Expectedly ICC results confirmed the up-regulation of P53 proteins in treated samples. Conclusion: Our results indicate the cytotoxicity of hypericin on Huh7 cells by affecting the expression of the gate keeper gene P53; furthermore it is suggested that this herb can be utilized simultaneously with modalities targeting P53 up-regulation or related molecular pathways. [GMJ.2020;9:e1896]


2020 ◽  
Author(s):  
Jikang Yang ◽  
Zhiyuan Xing

Abstract Background: Drug resistance is still a major obstacle during the cisplatin-based chemotherapy of hepatocellular carcinoma (HCC). Recently, studies have indicated that the population of CD133+ cancer cells is partially responsible for the failure of cancer treatment. However, the potential mechanisms are still unclear.Methods: CD133+ HepG2 and Huh7 cells were sorted via flow cytometry. CCK-8 assay was used to detect the cytotoxicity of cisplatin and silibinin against HCC cells. Western blot assay was performed to detect the protein expression, cleavage of caspases and release of cytochrome c from mitochondria into cytosol. Flow cytometry analysis was used to measure the apoptotic rate of CD133+ HepG2 and Huh7 cells.Results: CD133+ HepG2 and Huh7 cells were observed to exhibit obvious resistance against cisplatin. However, co-treatment with silibinin significantly reduced the cisplatin resistance of CD133+ HepG2 and Huh7 cells. Furthermore, although CD133+ HepG2 and Huh7 cells were resistant to cisplatin-induced apoptosis, co-treatment with silibinin enhanced the cisplatin-induced apoptosis through promoting the release of cytochrome c from mitochondria into cytosol. In the mechanism research, we proved that silibinin inhibited the expression of OPA1 in CD133+ HepG2 and Huh7 cells. Under the stress of cisplatin, silibinin promoted the collapse of mitochondria and increased the release of cytochrome c. As a result, caspases-dependent apoptosis was induced in CD133+ HepG2 and Huh7 cells which were co-treated with cisplatin and silibinin.Conclusion: Silibinin sensitizes CD133+ HCC cells to cisplatin-induced apoptosis through suppression of OPA1.


2021 ◽  
Author(s):  
Liyuan Hao ◽  
Yinglin Guo ◽  
Qing Peng ◽  
Zhiqin Zhang ◽  
Shenghao Li ◽  
...  

Abstract Hepatocellular carcinoma (HCC) was one of the most malignant cancers in the world. Cisplatin (DDP) was one of the main chemotherapy drugs for HCC, but the mechanism of DDP treatment for HCC remains unclear. In this presentation, we found that DDP inhibited the growth of HCC cells and promoted the expression of PD-1 and its ligand PD-L1 in cancer cells. Meanwhile, flow cytometry analysis revealed that DDP enhanced PD-1-CD8+ T cells expression and decreased PD-1+CD8+ T cells expression. ELISA analysis suggested that DDP decreased TGF-β expression in vivo. Therefore, the study indicated that DDP enhanced PD-1 and PD-L1 expression and inhibited the growth of HCC.


2020 ◽  
Vol 20 (3) ◽  
pp. 1292-1298
Author(s):  
Bing Wang ◽  
Wang-Xun Jin ◽  
Yun-Li Zhang ◽  
Ling Huang ◽  
Hai-Bin Ni ◽  
...  

Background: Hepatocellular carcinoma is one of the most common malignant tumors found all over the globe. Despite advances in surgery and chemotherapy, the five-year survival rate of patients with hepatocellular carcinoma is still low. It is known that the proliferation of hepatocellular carcinoma cells is closely related to the occurrence, development and prog- nosis of hepatocellular carcinoma. The present work investigates the expression of microRNA-489 (miR-489) in human hepatocellular carcinoma cells and its effect on the biological behavior of human hepatocellular carcinoma cells. Methods: The expression of miR-489 by fluorescence quantitative PCR detection in 30 patients with hepatoblastoma of liver cancer tissues and adjacent tissues was studied. Also, the determination of hepatoblastoma in four cell lines with differ- ent metastatic potential (HR8348, HCT116, HT29 and HEPG2) and the expression of miR-489 during miR-489 simulation process was studied. MTT assay, flow cytometry and Western blot analysis were performed to know the cell proliferation to detect the changes in cell cycle, apoptosis of cells, and SOX4 gene expression respectively. Results: RT-PCR results showed that the cells compared with pre-cancerous tissue, the expression level of miR-489 in hepatocellular carcinoma tissues than in adjacent tissue significantly decreased (P<0.05), and with liver cancer cell metastasis increased (P<0.05); analogue transfection constructed miR-489 overexpressing HEPG2 cell line by microRNA. MTT results showed that miR-489 can inhibit the proliferation of HEPG2 cells, the differences were statistically significant (P<0.05); flow cytometry results showed that miR-489 mimics was transfected into HEPG2 cells at 48 hours had no significant effect on cell cycle distribution (P > 0.05); but miR-489 expression could induce apoptosis, compared with the control group, the apoptosis of miR-489 mimics was significantly increased and the difference was statistically significant (P < 0.05). Conclusion: In conclusion, miR-489 can significantly inhibit the occurrence and development of hepatocellular carcinoma cells. The mechanism may be down regulated by the expression of SOX4 and inhibit cell proliferation. Further this study showed that the tumor cells SOX4 gene as a regulatory factor target the genes of miR-489 in hepatocellular carcinoma. Keywords: Hepatocellular carcinoma; mircroRNA-489; SOX4; apoptosis.


2017 ◽  
Vol 43 (5) ◽  
pp. 1803-1812 ◽  
Author(s):  
Zhijia Cao ◽  
Huainian Zhang ◽  
Xiaoyan Cai ◽  
Wei Fang ◽  
Dong Chai ◽  
...  

Background/Aims: Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and is a leading cause of cancer-related death worldwide. Luteolin, a flavonoid from traditional Chinese medicine, shows anti-cancer activity in many cancer cells, including HCC. However, the mechanism underlying the action of luteolin in HCC, especially its role in regulating cell autophagy, remains unclear. In the present study, we investigated the role of luteolin in regulating cell autophagy and the role of autophagy in luteolin-induced apoptosis. Methods: The 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay (MTT) was used to investigate cell viability. Flow cytometry analysis was used to detect the cell cycle and cell apoptosis. Hoechst 33342 staining was used to detect cell apoptosis. Transmission electron microscopy was used to investigate autophagy. qRT-PCR and western blotting were used to detect apoptosis- and autophagy-related mRNAs and proteins. Results: Luteolin reduced the viability of SMMC-7721 cells in a time and dose-dependent manner, and induced significant G0/G1-phase arrest. In addition, the results of flow cytometry analysis and Hoechst 33342 staining showed that luteolin treatment increased the number of apoptotic cells obviously, and the results of qRT-PCR and western blotting showed that luteolin treatment increased caspase 8 and decreased bcl-2 at the mRNA and protein levels. Furthermore, luteolin increased the number of intracellular autophagosomes, promoted LC3B-I conversion to LC3B-II, and increased Beclin 1 expression. Finally, co-treatment with the autophagy inhibitor chloroquine weakened the effects of luteolin on cell apoptosis. Conclusion: Luteolin induced apoptosis in human liver cancer SMMC-7721 cells, partially via autophagy. Thus, luteolin could be used as a regulator of autophagy in HCC treatment.


2021 ◽  
Author(s):  
Bijoya Sen ◽  
Savera Aggarwal ◽  
Rhisita Nath ◽  
Rashi Sehgal ◽  
Archana Rastogi ◽  
...  

AbstractPresence of dysfunctional senescent hepatocyte is a hallmark feature of cirrhosis. We now report the presence of senescent hepatocytes (p21 and p53 positive) in vicinity of infiltrated immune cells in hepatocellular carcinoma. Hence, we checked if senescent cells can alter fate of macrophage polarization and neutrophil extracellular trap (NETs) formation. Using an in vitro assay, senescence was induced in hepatoma cells (HepG2 and Huh7 cells) by doxorubicin treatment and senescent cell showed secretory phenotype with strong expression of cytokines (IL1β, IL6, IL8 and IL13) as evaluated by Flow cytometry. The senescent secretome from hepatoma cells induced macrophage differentiation predominantly with M2 markers (CD80, CD86) while that of non-senescent cell induced M1 phenotype (CD163, CD206) as analysed by flow cytometry. Human hepatocellular carcinoma harbouring senescent hepatocytes showed presence of M2 macrophages, while M1 macrophages were predominant in non-tumorous region. Additionally, the senescent secretome from Huh7 cells (p53mut) enhanced the NETs formation, while HepG2 (p53+/+) secretome had an inhibitory affect In conclusion, the “pro-inflammatory” senescent secretome drives non-inflammatory type M2 macrophage polarization and modulate neutrophil traps thereby modulating the microenvironment towards tumor promotion. Targeting senescent hepatocyte secretome appears a promising therapeutic target in liver cancer in future.Abstract FigureWork Highlight (Diagrammatic Representation).


2017 ◽  
Vol 35 (4_suppl) ◽  
pp. 324-324 ◽  
Author(s):  
Mehmet Akce ◽  
Luis M Vence ◽  
Jorge M Blando ◽  
Padmanee Sharma ◽  
Manal Hassan ◽  
...  

324 Background: Fibrolamellar hepatocellular carcinoma (FLHCC) is a rare variant of hepatocellular carcinoma (HCC) with about 200 annual cases reported globally. Surgical resection is the main treatment. Although FLHCC develops in the absence of cirrhosis surrounding hepatic parenchyma may have mononuclear cell and lymphocyte infiltrates. Immunotherapy by targeting programmed death 1 (PD-1) has emerged as a potential therapy for HCC and multiple clinical trials are being conducted, however little is known about immunological features of FLHCC. We aimed to analyze tumor microenvironment by immunohistochemistry and flow cytometry. Methods: Surgical samples of two FLHCC cases, 23 year old woman (case 1) and 25 year old man (case 2) who underwent neoadjuvant chemotherapy with 5 Fluorouracil + Interferon followed by surgical resection were analyzed. Expression of CD4, CD8, PD-1, programmed death ligand 1(PD-L1), CD45RO, CD 57, CD68, OX 40, inducible costimulator (ICOS), FoxP3, Gr-B was evaluated by immunohistochemistry. Flow cytometry analysis of CD8, CD4 effector (Teff) and regulatory (Treg) T cells were performed. Immune monitoring studies were conducted by the Immunotherapy Platform at MD Anderson Cancer Center. Results: By immunohistochemistry,expression of PD-1 and PDL-1 in the tumor sampleswas asfollows:PD-1 (5.9%) and PDL-1 (4.1%) of total cells in case 1 versus PD-1 (6.3%) and PDL-1 (3.5%) in case 2. FoxP3 expression was 0.3% and 1%, whereas ICOS expression was 0.2% and 1% in case 1 and 2, respectively. By flow cytometry analysis, case 1 presented the following frequencies: CD8+CTLA4+ (40%), CD8+PD1+ (40%) of total CD8 T cells, CD4+TeffCTLA4+ (55%), CD4+TeffPD1+ (30%), CD4+TeffICOS+ (15%) of total CD4 Teff cells and CD4+TregCTLA+ (90%), CD4+TregPD1+ (30%), CD4+TregICOS+ (70%) of total CD4 Tregs. Similarly, in case 2 the percentages were CD8+CTLA-4+ (20%), CD8+PD1+ (40%), CD4+TeffCTLA4+ (25%), CD4+TeffPD1+ (65%), CD4+TeffICOS+ (30%), CD4+TregCTLA+ (90%), CD4+TregPD1+ (70%), CD4+TregICOS+ (80%). Conclusions: PD-1, PDL-1, CTLA-4 and ICOS expression in FLHCC might render support to consideration of potential utilization of antibodies against PD-1, PDL-1 and CTLA-4 for treatment for a devastating cancer with unmet need for treatment.


2020 ◽  
Author(s):  
Olivia J. Kelada ◽  
Nicholas T. Gutsche ◽  
Meghan Bell ◽  
Rose M. Berman ◽  
Kwamena E. Baidoo ◽  
...  

BackgroundHepatocellular carcinoma (HCC) is the fifth most common cancer worldwide. While conventional imaging approaches like ultrasound, CT, and MRI play critical roles in the diagnosis and surveillance of HCC, improved methods for detection and assessment of treatment response are needed. One promising approach is the use of radiolabeled antibodies for positron emission tomography (immunoPET) imaging. Glypican-3 (GPC3) is a proteoglycan that is highly expressed in the majority of HCC tumors. GPC3-specific antibodies are used to diagnose HCC histopathologically, and have been proposed as a treatment of HCC. Here, we design, synthesize and demonstrate that our humanized immunoPET agent, [89Zr]Zr-DFO-TAB-H14, can stoichiometrically bind to models of human liver cancer with varied GPC3 expression. Methods: The GPC3-specific monoclonal humanized IgG1, TAB-H14, was used as a scaffold for engineering our immunoPET agent. Fluorescent and deferroxamine (DFO) chelate conjugates of TAB-H14 were characterized using mass spectrometry. Binding affinity of TAB-H14 and conjugates for GPC3 was determined in cell-free biolayer interferometry, and cell-based radioimmunoassays. GPC3-expression was assessed by flow cytometry and immunofluorescence using commercially available anti-GPC3 antibodies and TAB-H14 in GPC3−(A431) and GPC3+ cell lines including an engineered line (A431-GPC3+, G1) and liver cancer lines (HepG2, Hep3B, and Huh7). DFO-TAB-H14, was radiolabeled with Zr-89. Mice were subcutaneously engrafted with the aforementioned cell lines and in vivo target engagement of the immunoPET agent [89Zr]Zr-DFO-TAB-H14 was determined using PET/CT, quantitative biodistribution, and autoradiography. Results: TAB-H14 demonstrated subnanomolar to nanomolar affinity for human GPC3. Fluorescently tagged TAB-H14 was able to bind to GPC3 on cell membranes of GPC3-expressing lines by flow cytometry. These results were confirmed by immunofluorescence staining of A431, G1 HepG2, Hep3B, and Huh7 tumor sections. ImmunoPET imaging with [89Zr]Zr-DFO-TAB-H14 showed stoichiometric tumor uptake corresponding to the cell surface expression levels. Autoradiography and immunostaining confirmed in vivo findings. Conclusion: We systematically demonstrate that the humanized immnoPET agent [89Zr]Zr-DFO-TAB-H14 specifically and stoichiometrically binds to GPC3 in several models of human liver cancer, serving as a promising in vivo GPC3 sensor. This agent may provide utility in HCC diagnosis and surveillance, and the selection of candidates for GPC3-directed therapies.


2011 ◽  
Vol 5 (3) ◽  
pp. 153-160 ◽  
Author(s):  
Giuseppe Carruba ◽  
Vitale Miceli ◽  
Letizia Cocciadiferro ◽  
Maurizio Zarcone ◽  
Biagio Agostara ◽  
...  

AbstractBackground:We investigated aromatase (Aro)-driven estrogen formation in non-tumoral and malignant liver tissues and cells, also in relation to expression of the estrogen receptors α and β (ERα and ERβ) and amphiregulin (AREG), aiming to gain insights into the potential role of estrogens in human hepatocellular carcinoma (HCC).Materials and methods:Chromatographic and reverse transcriptase polymerase chain reaction (RT-PCR) analyses were used to assess activity and expression of the Aro enzyme and AREG as well as the expression of wild-type and variant ERs, both in vivo and in vitro.Results:Following 24 h and 72 h incubation of liver tissues or cells with testosterone, human HCC tissues and HepG2 hepatoma cells showed elevated Aro activity (estrogen formation, respectively, of 20% and 52%–99%). By contrast, no Aro activity could be detected in non-tumoral tissues and HA22T liver cancer cells. Cirrhotic samples and Huh7 cells exhibited intermediate enzyme activity, with estrogen formation of 4% and 34%, respectively. Markedly lower or undetectable Aro mRNA levels were observed in HA22T cells and non-tumoral liver tissues compared with HepG2 cells and HCC samples. Cirrhotic specimens displayed variable transcript levels. Interestingly, no or low expression of wild-type ERα and ERβ could be observed in liver cancer cells and malignant tissues. However, ubiquitous expression of the hERα46 variant and occasional expression of the hERβ2/Cx variant were observed in cancer tissues and cells.Conclusions:It is noteworthy that the pattern of wild-type ERα was inversely related to Aro, whilst AREG expression was consistently associated with that of Aro. This combined evidence suggests that locally elevated Aro activity may increase malignant cell proliferation also through AREG signalling.


2020 ◽  
Author(s):  
Chen Fang ◽  
Yang Longfei ◽  
Wang Xiaofeng ◽  
Zheng Yang ◽  
Chen Jiaxi ◽  
...  

Abstract Purpose: To explore the regulatory role of HSF4 in hepatocellular carcinoma stemness property maintaining and analysis the clinical significance of HSF4 in hepatocellular carcinoma. Materials and methods: Tumor spheroid formation assay was conducted to assess stemness property and enrich hepatocellular carcinoma stem-like cells. qRT-PCR and Western Blot was used to detect genes mRNA and protein expression level. KM-plotter database wad used to analyze the correlation between HSF4 and overall survival (OS) in HCC patients. Result: mRNA level of HSF4, as well as stemness-associated genes, was significantly higher in hepatocellular carcinoma tissue then in adjacent normal tissue. Positive correlation between expression level of HSF4 and SOX (r=0.1668, p<0.05), NANOG (r=0.7, p<0.05), POUSF1(r=0.7362, p<0.05), CD44(r=0.0128, p<0.05) was observed. The KM plotter showed that there is no significant difference between HSF4 high patients and HF4 low patients in term of overall survival (OS)(p=0.48). However, significant difference in terms of progression-free survival (PFS)(p=0.019) and relapse-free survival (RFS)(p=0.005) between these two groups was observed. In vitro assay results also suggest the positive correlation between HSF4 and stemness-associated genes. Increased HSF4 expression confers HCC enhanced tumor spheroid formation ability. Conclusion: HSF4 is significantly increased in liver cancer stem-like cells, indicating the possible contribution of HSF4 to stemness properties maintenance in liver cancer stem-like cells. Silencing HSF4 inhibits proliferation of hepatoma cells.


Sign in / Sign up

Export Citation Format

Share Document