Ferroptosis-centered Disease Response Information
General Information of the Disease (ID: DIS00031)
Name |
Hepatocellular carcinoma
|
||||
---|---|---|---|---|---|
ICD |
ICD-11: 2C12
|
Full List of Target(s) of This Ferroptosis-centered Disease
Stearoyl-CoA desaturase (SCD)
In total 3 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Lactate | Investigative | |||
Responsed Regulator | Hydroxycarboxylic acid receptor 1 (HCAR1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
AMPK signaling pathway | hsa04152 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | CAF cells | Normal | Carassius auratus | CVCL_R883 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Female mice aged around 6-7 weeks were used for this study, which were purchased through Laboratory Animal Center of Chongqing Medical University from Vital River Co. Ltd (Beijing, China).After one week, each mouse was injected subcutaneously with 100 uL of Huh-7 cell suspension (5 x 106 units) to establish the tumor model. The mice were grouped randomly, and then subjected to different treatments after subcutaneous tumors became visually detectable.
Click to Show/Hide
|
||||
Response regulation | Lactate regulates the ferroptosis of hepatocellular carcinoma cells. And blocking the lactate uptake via hydroxycarboxylic acid receptor 1 (HCAR1)/MCT1 inhibition promotes ferroptosis by activating the AMPK to downregulate SCD1, which may synergize with its acyl-coenzyme A synthetase 4 (ACSL4)-promoting effect to amplify the ferroptotic susceptibility. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Lactate | Investigative | |||
Responsed Regulator | Sterol regulatory element-binding protein 1 (SREBF1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
AMPK signaling pathway | hsa04152 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | CAF cells | Normal | Carassius auratus | CVCL_R883 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Female mice aged around 6-7 weeks were used for this study, which were purchased through Laboratory Animal Center of Chongqing Medical University from Vital River Co. Ltd (Beijing, China).After one week, each mouse was injected subcutaneously with 100 uL of Huh-7 cell suspension (5 x 106 units) to establish the tumor model. The mice were grouped randomly, and then subjected to different treatments after subcutaneous tumors became visually detectable.
Click to Show/Hide
|
||||
Response regulation | The monocarboxylate transporter 1 (MCT1)-mediated lactate uptake could promote ATP production in hepatocellular carcinoma (HCC) cells and deactivate the energy sensor AMP-activated protein kinase (AMPK), leading to the upregulation of SREBP1 (SREBF1) and the downstream stearoyl-coenzyme A (CoA) desaturase-1 (SCD1) to enhance the production of anti-ferroptosis monounsaturated fatty acids. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [16] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Pathway Response | Glutathione metabolism | hsa00480 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
In Vivo Model |
Six-week-old male BALB/c athymic nude mice were purchased from the Experimental Animal Center of Peking (Beijing, China). Stable cells (5 x 106) were seeded into the right flanks of the mice. After the xenografts had grown to 200 mm3, saline as a vehicle or sorafenib (30 mg/kg) was administered by gavage every day, and the mice were euthanized by the cervical dislocation method five weeks later. Before sacrifice, the tumor sizes and body weights were measured twice per week. The tumor volume (V) was calculated as follows: (L x W2)/2 (length, L, and width, W). The xenografts were excised and further assessed.
Click to Show/Hide
|
||||
Response regulation | Sorafenib decreased HBXIP expression, and overexpression of HBXIP blocked sorafenib-induced Hepatocellular carcinoma cell death. Regarding the molecular mechanism, HBXIP transcriptionally induced the expression of stearoyl-CoA desaturase (SCD) via coactivating the transcriptional factor ZNF263, resulting in the accumulation of free fatty acids and suppression of ferroptosis. | ||||
Phospholipid hydroperoxide glutathione peroxidase (GPX4)
In total 29 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [2] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | Ketamine | Investigative | |||
Responsed Regulator | PVT1 (IncRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
BALB/c nude mice (age 6 weeks) were brought from the Laboratory Animal Center of Chinese Academy of Sciences (China). HepG2 cell suspension (100 uL, 5 x 105 per site) was hypodermically inoculated into the fat pad of mice. Tumor volume was calculated as follows: tumor volume (mm3) = 0.5 x width (mm)2 x length (mm). When tumor size reached 100 mm3, mice were treated with ketamine (20 mg/kg) or saline intraperitoneally. The mice were succumbed to death when tumor size reached 1000 mm3. Tumors were isolated and weighted. All animal experiments were carried out in accordance with the National Institutes of Health guide for the care and use of Laboratory animals (NIH Publications No. 8023, revised 1978).
Click to Show/Hide
|
||||
Response regulation | LncPVT1 directly interacted with miR-214-3p to impede its role as a sponge of GPX4. Depletion of lncPVT1 accelerated the ferroptosis of liver cancer cells, whereas miR-214-3p inhibition and GPX4 overexpression reversed this effect. In this work, we determined that ketamine suppressed viability of liver cancer cells and induced ferroptosis and identified the possible regulatory mechanism of lncPVT1/miR-214-3p/GPX4 axis. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [2] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | Ketamine | Investigative | |||
Responsed Regulator | hsa-miR-214-3p (miRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
BALB/c nude mice (age 6 weeks) were brought from the Laboratory Animal Center of Chinese Academy of Sciences (China). HepG2 cell suspension (100 uL, 5 x 105 per site) was hypodermically inoculated into the fat pad of mice. Tumor volume was calculated as follows: tumor volume (mm3) = 0.5 x width (mm)2 x length (mm). When tumor size reached 100 mm3, mice were treated with ketamine (20 mg/kg) or saline intraperitoneally. The mice were succumbed to death when tumor size reached 1000 mm3. Tumors were isolated and weighted. All animal experiments were carried out in accordance with the National Institutes of Health guide for the care and use of Laboratory animals (NIH Publications No. 8023, revised 1978).
Click to Show/Hide
|
||||
Response regulation | LncPVT1 directly interacted with miR-214-3p to impede its role as a sponge of GPX4. Depletion of lncPVT1 accelerated the ferroptosis of liver cancer cells, whereas miR-214-3p inhibition and GPX4 overexpression reversed this effect. In this work, we determined that ketamine suppressed viability of liver cancer cells and induced ferroptosis and identified the possible regulatory mechanism of lncPVT1/miR-214-3p/GPX4 axis. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [3] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocarcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Oxaliplatin | Investigative | |||
Responsed Regulator | LINC01134 (IncRNA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Response regulation | LINC01134 was positively correlated with GPX4 or Nrf2, demonstrating the clinical significance of LINC01134, Nrf2 and GPX4 in OXA resistance of hepatocellular carcinoma (HCC). Silenced LINC01134 enhances Oxaliplatin sensitivity by facilitating ferroptosis through GPX4 in hepatocarcinoma. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [4] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Punicalin | Investigative | |||
Responsed Regulator | L-seryl-tRNA(Sec) kinase (PSTK) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
SNU-387 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0250 | ||
SNU-182 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0090 | ||
SNU-398 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0077 | ||
WRL 68 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0581 | ||
HUVECs (Human umbilical vein endothelial cells) | |||||
JHH-2 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_2786 | ||
JHH-7 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_2805 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Li-7 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_3840 | ||
In Vivo Model |
Female Nod-SCID mice of 6-8 weeks old were purchased from HFK BIOSCIENCE (Beijing). Hep3B-vehicle/Hep3B-PSTK-KO cells were harvested and injected subcutaneously (1 x 107 cells in 200 uL PBS) into Nod-SCID mice (upper flank). Treatments were started when tumor volumes reached around 50 mm3. Included mice were randomly divided into four groups and injected intraperitoneally with Abemaciclib (50 mg/kg, every other day) or vehicle. Mice were sacrificed when the tumor volume exceeded 2000 mm3. PSTK-KO or vehicle Hep3B cells were implanted and treated with Sorafenib (50 mg/kg, every other day) or Erastin (50 mg/kg, every other day) for 42 days. Tumor volumes were monitored and quantified by the modified ellipsoidal formula, tumor volume = (length x width2)/2. To check the efficacities and appraisal the side effects of PSTK inhibitors, Hep3B cells were harvested and in injected subcutaneously (5 x 106 cells in 200 uL PBS) into Nod-SCID mice (upper flank). Treatments were started when tumor volumes reached around 50 mm3. Included mice were randomly divided into six groups and intragastrically treated with Punicalin (100 mg/kg, every day), Geraniin (100 mg/kg, every day), Sorafenib (50 mg/kg, every day) with or without PSTK inhibitors (Punicalin/Geraniin) for 30 days. Tumor volumes and mice weights were measured every three days.
Click to Show/Hide
|
||||
Response regulation | The depletion of PSTK resulted in the inactivation of glutathione peroxidative 4 (GPX4) and the disruption of glutathione (GSH) metabolism owing to the inhibition of selenocysteine and cysteine synthesis, thus enhancing the induction of ferroptosis upon targeted chemotherapeutic treatment. Punicalin, an agent used to treat hepatitis B virus (HBV), was identified as a possible PSTK inhibitor that exhibited synergistic efficacy when applied together with Sorafenib to treat Hepatocellular carcinoma in vitro and in vivo. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Responsed Regulator | Protein lifeguard 4 (TMBIM4) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
In Vivo Model |
To generate murine subcutaneous tumours, 1 x 107 control shRNA or S1R-knockdown Huh7 cells in 200 uL of PBS were injected subcutaneously to the right of the dorsal midline. At day seven, the mice were randomly divided into groups and treated with sorafenib (10 mg/kg/intraperitoneal injection (i.p.), once every other day) for 2 weeks. On day 28, tumours were removed.
Click to Show/Hide
|
||||
Response regulation | S1R (TMBIM4) protects hepatocellular carcinoma cells against sorafenib and subsequent ferroptosis. Inhibition of S1R by RNAi and antagonists markedly increased the anticancer activity of sorafenib by modulating the expression of GPX4, iron metabolism and ROS. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [6] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Responsed Regulator | HCG18 (IncRNA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model | SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
HCCLM3 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_6832 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
BALB/c nude mice (4-6 weeks old) from Beijing Vital River Laboratory Animal Technology (Beijing, China) were reared in a standard laboratory with free access to food and water. Lentivirus LV-sh-NC and LV-sh-HCG18 were from GenePharma (Shanghai, China). In order to establish subcutaneous xenograft tumor models, Huh7-SR cells were infected with lentivirus LV-sh-NC or LV-sh-HCG18 and then resuspended in PBS at 5 x 105/mL. Totally 100 uL cells were subcutaneously injected into the right dorsal area of each nude mouse. When the tumor volume reached 150 mm3, sorafenib (10 mg/kg) was orally administered to nude mice once a day to the end. Tumor volume (V) was calculated: V = 0.5 x L x W2, where L and W were defined as tumor length (L) and width (W). After 28 days of cell injection, the nude mice were euthanized by intraperitoneal injection of excessive pentobarbital sodium (100 mg/kg).
Click to Show/Hide
|
||||
Response regulation | HCG18 sponged miR-450b-5p to regulate GPX4. Collectively, Silencing HCG18 inhibits GPX4 by binding to miR-450b-5p, promotes GPX4-inhibited ferroptosis, and averts sorafenib resistance in hepatocellular carcinoma (HCC). Silencing HCG18 inhibited cell proliferation, promoted apoptosis, and impaired sorafenib resistance. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [21] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | 2-pyridylhydrazone dithiocarbamate s-acetic acid | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
Cell apoptosis | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | 2-pyridylhydrazone dithiocarbamate s-acetic acid (PdtaA) induced both apoptosis and cell cycle arrest. Notably, PdtaA also induced ferroptosis via downregulation of GPx4 and xCT in liver cancer cells. Autophagy inhibitor 3-methyladenin or genetic knockdown of NCOA4 was employed to inhibit ferritinophagy, which significantly neutralized the action of PdtaA in both apoptosis and ferroptosis. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [18] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Atractylodin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hccm (Human hepatocellular carcinoma cells) | |||||
Response regulation | Atractylodin can inhibit the proliferation, migration, and invasion of Huh7 and Hccm liver cancer cells, and induce cell apoptosis and cell cycle arrest. In addition, atractylodin may induce ferroptosis in hepatocellular carcinoma cells by inhibiting the expression of GPX4 and FTL proteins, and up-regulating the expression of ACSL4 and TFR1 proteins. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [22] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Auranofin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Response regulation | Auranofin/buthionine sulfoxime (BSO) and Erastin/BSO cotreatment alters redox homeostasis by increasing levels of Nrf2 and HO-1 and decreasing GPX4 levels. Targeting these two main ferroptotic pathways simultaneously can overcome chemotherapy resistance in hepatocellular carcinoma (HCC). | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [22] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Erastin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Response regulation | Auranofin/buthionine sulfoxime (BSO) and Erastin/BSO cotreatment alters redox homeostasis by increasing levels of Nrf2 and HO-1 and decreasing GPX4 levels. Targeting these two main ferroptotic pathways simultaneously can overcome chemotherapy resistance in hepatocellular carcinoma (HCC). | ||||
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target | [22] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | l-Buthionine sulfoximine | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Response regulation | Auranofin/buthionine sulfoxime (BSO) and Erastin/BSO cotreatment alters redox homeostasis by increasing levels of Nrf2 and HO-1 and decreasing GPX4 levels. Targeting these two main ferroptotic pathways simultaneously can overcome chemotherapy resistance in hepatocellular carcinoma (HCC). | ||||
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target | [23] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Seco-Lupane Triterpene Derivatives | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | A new seco-lupane triterpene derivative, compound21, was found to regulate cell growth through the cell cycle and ferroptosis, which in turn inhibited the proliferation, migration, and invasion of HepG2 cells. And it was found that compound 21 significantly upregulated ACSL4 protein expression and downregulated GPX4 protein expression. It has the potential to become an effective new drug for the treatment of hepatocellular carcinoma. | ||||
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target | [24] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Solasonine | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
HepaRG cells | Hepatocellular carcinoma | Homo sapiens | CVCL_9720 | ||
In Vivo Model |
BALB/c nude mice aged 4-6 weeks, weighing 15~20 g, were purchased from Shanghai SLAC Laboratory Animal Co.,Ltd (Shanghai, China). Following acclimation, the right flank of each experimental mouse was subcutaneously injected with HepG2 cells (2 x 106) suspended in PBS (200 uL) and then randomly assigned to: (i) the control group and received no further treatment or (ii) the intervention group and received solasonine (50 mg/kg body weight) in an equal volume of PBS. Tumor volumes were measured every 5 days. After 30 days, the mice were sacrificed and the tumors were resected, weighed, and processed for histological analysis.
Click to Show/Hide
|
||||
Response regulation | Solasonine increased lipid ROS levels in HepG2 cells by suppression of GPX4 and GSS. However, the use of a ferroptosis inhibitor reversed solasonine-induced ROS production and cell apoptosis. Taken together, solasonine promotes ferroptosis of hepatocellular carcinoma cells via GPX4-induced destruction of the glutathione redox system. | ||||
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target | [20] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | Peroxisome proliferator-activated receptor alpha (PPARA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hepa 1-6 cells | Hepatocellular carcinoma | Mus musculus | CVCL_0327 | |
In Vivo Model |
C57BL/6J SPF mice were purchased from Huazhong Agricultural University Experimental Animal Center. Mice were given tertian intraperitoneal injections of either PBS (control) or dextriferron (500 mg/kg body weight) for 2 weeks and then sacrificed. Mice were given a daily intraperitoneal injection of either vehicle or ferrostatin-1 (Fer1, 1 mg/kg body weight) for 3 weeks before sacrificed.
Click to Show/Hide
|
||||
Response regulation | PPARa activation alleviates iron overload-induced ferroptosis in mouse livers through Gpx4 and TRF, suggesting that PPAR may be a promising therapeutic target for drug discovery in ferroptosis-related tissue injuries in Hepatocellular carcinoma. | ||||
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target | [25] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Maleylacetoacetate isomerase (GSTZ1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
SNU-449 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0454 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Mice were divided into five groups as follows: WT + DMSO (control), WT + Sora, Gstz1-/-+DMSO, Gstz1-/-+Sora, and Gstz1-/-+Sora + RSL3. Each group included three male and three female mice. At 2 weeks of age, all mice were administered an intraperitoneal injection of diethylnitrosamine (DEN; Sigma, St. Louis, MO, USA) at a dose of 75 mg/kg. At the third week, the mice were intraperitoneally administered carbon tetrachloride (CCl4; Macklin, Shanghai, China) at 2 ml/kg twice a week for 12 weeks. In the WT + Sora and Gstz1-/-+Sora group, the mice at 22 weeks were administered intraperitoneally sorafenib (30 mg/kg) every 2 days for 4 weeks until euthanasia. In the Gstz1-/-+Sora + RSL3 group, in addition to sorafenib administration as described above, the mice were injected intraperitoneally with RSL3 (10 mg/kg) every 2 days for 4 weeks at the same weeks.
Click to Show/Hide
|
||||
Response regulation | GSTZ1 enhanced sorafenib-induced ferroptosis by inhibiting the NRF2/GPX4 axis in hepatocellular carcinoma (HCC) cells. Combination therapy of sorafenib and GPX4 inhibitor RSL3 may be a promising strategy in HCC treatment. | ||||
Experiment 16 Reporting the Ferroptosis-centered Disease Response by This Target | [26] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Glucose-6-phosphate 1-dehydrogenase (G6PD) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | WRL 68 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0581 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SNU-387 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0250 | ||
In Vivo Model |
Animal experiments were conducted under the guidance of Animal Management Regulations in Chongqing University. The tumor volume was calculated as follows: (length x width2)/2. After 24 days, the mice were killed and their tumors were collected, fixed and sectioned, stained by hematoxylin and eosin, and examined by a light microscopy for histological changes.
Click to Show/Hide
|
||||
Response regulation | G6PD (glucose-6-phosphate dehydrogenase) was highly expressed in hepatocellular carcinoma and was associated with poor prognosis. G6PD promoted the proliferation, migration and invasion, as well as inhibited ferroptosis in HCC cells. G6PD inhibited ferroptosis inin HCC cells through POR. GPX4 was positively regulated by G6PD. | ||||
Experiment 17 Reporting the Ferroptosis-centered Disease Response by This Target | [26] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | NADPH--cytochrome P450 reductase (POR) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | WRL 68 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0581 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SNU-387 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0250 | ||
In Vivo Model |
Animal experiments were conducted under the guidance of Animal Management Regulations in Chongqing University. The tumor volume was calculated as follows: (length x width2)/2. After 24 days, the mice were killed and their tumors were collected, fixed and sectioned, stained by hematoxylin and eosin, and examined by a light microscopy for histological changes.
Click to Show/Hide
|
||||
Response regulation | G6PD (glucose-6-phosphate dehydrogenase) was highly expressed in hepatocellular carcinoma and was associated with poor prognosis. G6PD promoted the proliferation, migration and invasion, as well as inhibited ferroptosis in HCC cells. G6PD inhibited ferroptosis inin HCC cells through POR. GPX4 was positively regulated by G6PD. | ||||
Experiment 18 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Cyclic AMP-dependent transcription factor ATF-4 (ATF4) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
Response regulation | The expression of ferroptosis-related protein GPX4 decreased after HULC knockdown, and the GPX4 expression level was reversed when the inhibitor miR-3200-5p was added simultaneously. HULC was found to function as a ceRNA of miR-3200-5p, and miR-3200-5p regulates ferroptosis by targeting ATF4, resulting in the inhibition of proliferation and metastasis within hepatocellular carcinoma cells. | ||||
Experiment 19 Reporting the Ferroptosis-centered Disease Response by This Target | [28] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | RNA-binding motif, single-stranded-interacting protein 1 (RBMS1) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
C57BL/6 mice (4- to 6-week-old male) were fed in a pathogen-free vivarium under standard conditions at the animal care facility at Sun Yat-sen University. Hepa 1-6 cells transduced with RBMS1 or GPX4 or circIDE overexpression lentiviral vectors were subcutaneously injected into the right flank of mice in 100 ul of sterile PBS. IVIS images were taken.
Click to Show/Hide
|
||||
Response regulation | RBMS1 overexpression inhibited hepatocellular Carcinoma (HCC) cell growth by attenuating the expression of glutathione peroxidase 4 (GPX4)and further facilitated ferroptosis in vitro and in vivo. More importantly, a novel circIDE (hsa_circ_0000251) was identified to elevate RBMS1 expression via sponging miR-19b-3p in HCC cells. | ||||
Experiment 20 Reporting the Ferroptosis-centered Disease Response by This Target | [29] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Cholangiocarcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | Transcription factor JunD (JUND) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | hIBECs (Human intrahepatic biliary epithelial cells) | ||||
HuCC-T1 cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_0324 | ||
HCCC-9810 cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_6908 | ||
QBC939 cells | Cholangiocarcinoma | Homo sapiens | CVCL_6942 | ||
HuH-28 cells | Cholangiocarcinoma | Homo sapiens | CVCL_2955 | ||
RBE cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_4896 | ||
In Vivo Model |
For the proliferation assays, HuCCT1 cells with linc00976 knockdown, linc00976 overexpression, and negative control were subcutaneously injected into BALB/c nude mice. The mice were weighed every week and euthanized 5 weeks after injection. Finally, tumors were dissected and weighed. For the proliferation assays, HuCCT1 cells with linc00976 knockdown, linc00976 overexpression, and negative control were subcutaneously injected into BALB/c nude mice. The mice were weighed every week and euthanized 5 weeks after injection. Finally, tumors were dissected and weighed.
Click to Show/Hide
|
||||
Response regulation | JUND promotes linc00976 transcription, and linc00976 plays a crucial role in accelerating Cholangiocarcinoma tumorigenesis and metastasis and inhibiting ferroptosis by modulating the miR-3202/GPX4 axis. | ||||
Experiment 21 Reporting the Ferroptosis-centered Disease Response by This Target | [28] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | hsa-miR-19b-3p (miRNA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
C57BL/6 mice (4- to 6-week-old male) were fed in a pathogen-free vivarium under standard conditions at the animal care facility at Sun Yat-sen University. Hepa 1-6 cells transduced with RBMS1 or GPX4 or circIDE overexpression lentiviral vectors were subcutaneously injected into the right flank of mice in 100 ul of sterile PBS. IVIS images were taken.
Click to Show/Hide
|
||||
Response regulation | RBMS1 overexpression inhibited hepatocellular Carcinoma (HCC) cell growth by attenuating the expression of glutathione peroxidase 4 (GPX4)and further facilitated ferroptosis in vitro and in vivo. More importantly, a novel circIDE (hsa_circ_0000251) was identified to elevate RBMS1 expression via sponging miR-19b-3p in HCC cells. | ||||
Experiment 22 Reporting the Ferroptosis-centered Disease Response by This Target | [30] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | hsa-miR-541-3p (miRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | THLE-2 cells | Normal | Homo sapiens | CVCL_3803 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
HCCLM3 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_6832 | ||
In Vivo Model |
The 5-week-old BALB/c male nude mice (n = 10) were purchased from the Animal Center of the Chinese Academy of Medical Sciences (Beijing, China). A number of 2 x 106 HuH-7 cells were transfected with lentiviral vectors containing sh-circIL4R or sh-NC to establish the stably expressed cell lines. Ten mice were subcutaneously injected with HuH-7 cells with sh-circIL4R or sh-NC (five mice per group), constructing the xenograft model of HCC in vivo. Every 5 days after injection, tumor size was measured by a vernier caliper and tumor volume (length x width2 x 0.5) was calculated.
Click to Show/Hide
|
||||
Response regulation | CircIL4R acted as a miR-541-3p sponge to regulate its target glutathione peroxidase 4 (GPX4). GPX4 upregulation relieved the miR-541-3p-induced tumor inhibition and ferroptosis aggravation. CircIL4R played an oncogenic role in hepatocellular carcinoma via the miR-541-3p/GPX4 axis in vivo. | ||||
Experiment 23 Reporting the Ferroptosis-centered Disease Response by This Target | [29] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Cholangiocarcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | hsa-miR-3202 (miRNA) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | hIBECs (Human intrahepatic biliary epithelial cells) | ||||
HuCC-T1 cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_0324 | ||
HCCC-9810 cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_6908 | ||
QBC939 cells | Cholangiocarcinoma | Homo sapiens | CVCL_6942 | ||
HuH-28 cells | Cholangiocarcinoma | Homo sapiens | CVCL_2955 | ||
RBE cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_4896 | ||
In Vivo Model |
For the proliferation assays, HuCCT1 cells with linc00976 knockdown, linc00976 overexpression, and negative control were subcutaneously injected into BALB/c nude mice. The mice were weighed every week and euthanized 5 weeks after injection. Finally, tumors were dissected and weighed. For the proliferation assays, HuCCT1 cells with linc00976 knockdown, linc00976 overexpression, and negative control were subcutaneously injected into BALB/c nude mice. The mice were weighed every week and euthanized 5 weeks after injection. Finally, tumors were dissected and weighed.
Click to Show/Hide
|
||||
Response regulation | JUND promotes linc00976 transcription, and linc00976 plays a crucial role in accelerating Cholangiocarcinoma tumorigenesis and metastasis and inhibiting ferroptosis by modulating the miR-3202/GPX4 axis. | ||||
Experiment 24 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | hsa-miR-3200-5p (miRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
Response regulation | The expression of ferroptosis-related protein GPX4 decreased after HULC knockdown, and the GPX4 expression level was reversed when the inhibitor miR-3200-5p was added simultaneously. HULC was found to function as a ceRNA of miR-3200-5p, and miR-3200-5p regulates ferroptosis by targeting ATF4, resulting in the inhibition of proliferation and metastasis within hepatocellular carcinoma cells. | ||||
Experiment 25 Reporting the Ferroptosis-centered Disease Response by This Target | [31] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | HEPFAL (IncRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
Response regulation | LncRNA HEPFAL promotes the ubiquitination of SLC7A11, resulting in a decrease in GSH production, which in turn affects the activity of GPX4 and ultimately leads to the occurrence of ferroptosis. And LncRNA HEPFAL has the potential as a target for the diagnosis and treatment of hepatocellular carcinoma. | ||||
Experiment 26 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | HULC (IncRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
Response regulation | The expression of ferroptosis-related protein GPX4 decreased after HULC knockdown, and the GPX4 expression level was reversed when the inhibitor miR-3200-5p was added simultaneously. HULC was found to function as a ceRNA of miR-3200-5p, and miR-3200-5p regulates ferroptosis by targeting ATF4, resulting in the inhibition of proliferation and metastasis within hepatocellular carcinoma cells. | ||||
Experiment 27 Reporting the Ferroptosis-centered Disease Response by This Target | [29] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Cholangiocarcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | LINC00976 (IncRNA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | hIBECs (Human intrahepatic biliary epithelial cells) | ||||
HuCC-T1 cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_0324 | ||
HCCC-9810 cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_6908 | ||
QBC939 cells | Cholangiocarcinoma | Homo sapiens | CVCL_6942 | ||
HuH-28 cells | Cholangiocarcinoma | Homo sapiens | CVCL_2955 | ||
RBE cells | Intrahepatic cholangiocarcinoma | Homo sapiens | CVCL_4896 | ||
In Vivo Model |
For the proliferation assays, HuCCT1 cells with linc00976 knockdown, linc00976 overexpression, and negative control were subcutaneously injected into BALB/c nude mice. The mice were weighed every week and euthanized 5 weeks after injection. Finally, tumors were dissected and weighed. For the proliferation assays, HuCCT1 cells with linc00976 knockdown, linc00976 overexpression, and negative control were subcutaneously injected into BALB/c nude mice. The mice were weighed every week and euthanized 5 weeks after injection. Finally, tumors were dissected and weighed.
Click to Show/Hide
|
||||
Response regulation | JUND promotes linc00976 transcription, and linc00976 plays a crucial role in accelerating Cholangiocarcinoma tumorigenesis and metastasis and inhibiting ferroptosis by modulating the miR-3202/GPX4 axis. | ||||
Experiment 28 Reporting the Ferroptosis-centered Disease Response by This Target | [30] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | CircIL4R (circRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | THLE-2 cells | Normal | Homo sapiens | CVCL_3803 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
HCCLM3 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_6832 | ||
In Vivo Model |
The 5-week-old BALB/c male nude mice (n = 10) were purchased from the Animal Center of the Chinese Academy of Medical Sciences (Beijing, China). A number of 2 x 106 HuH-7 cells were transfected with lentiviral vectors containing sh-circIL4R or sh-NC to establish the stably expressed cell lines. Ten mice were subcutaneously injected with HuH-7 cells with sh-circIL4R or sh-NC (five mice per group), constructing the xenograft model of HCC in vivo. Every 5 days after injection, tumor size was measured by a vernier caliper and tumor volume (length x width2 x 0.5) was calculated.
Click to Show/Hide
|
||||
Response regulation | CircIL4R acted as a miR-541-3p sponge to regulate its target glutathione peroxidase 4 (GPX4). GPX4 upregulation relieved the miR-541-3p-induced tumor inhibition and ferroptosis aggravation. CircIL4R played an oncogenic role in hepatocellular carcinoma via the miR-541-3p/GPX4 axis in vivo. | ||||
Experiment 29 Reporting the Ferroptosis-centered Disease Response by This Target | [28] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | CircIDE (circRNA) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
C57BL/6 mice (4- to 6-week-old male) were fed in a pathogen-free vivarium under standard conditions at the animal care facility at Sun Yat-sen University. Hepa 1-6 cells transduced with RBMS1 or GPX4 or circIDE overexpression lentiviral vectors were subcutaneously injected into the right flank of mice in 100 ul of sterile PBS. IVIS images were taken.
Click to Show/Hide
|
||||
Response regulation | RBMS1 overexpression inhibited Hepatocellular Carcinoma (HCC) cell growth by attenuating the expression of glutathione peroxidase 4 (GPX4)and further facilitated ferroptosis in vitro and in vivo. More importantly, a novel circIDE (hsa_circ_0000251) was identified to elevate RBMS1 expression via sponging miR-19b-3p in HCC cells. | ||||
Nuclear factor erythroid 2-related factor 2 (NFE2L2)
In total 13 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [7] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Tiliroside | Investigative | |||
Responsed Regulator | Serine/threonine-protein kinase TBK1 (TBK1) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
All animal studies were approved by the Committee on Ethics of Animal Experiments of Binzhou Medical University (approval no: BZMU-IACUC-2021-331, date: 09/10/2021). To generate the ectopic HCC mouse models, HepG2-luciferase cells (HepG2 cells transfected with luciferase gene) were suspended in serum-free media and matrigel (BD Biosciences) at a ratio of 1:1 v/v. A total of 2.5 x 106 HepG2-luciferase cells/100 ul were injected into the left axilla of mice. After reaching a tumor size of 100-150 mm3, all mice were randomly divided into four groups: control (vehicle, intraperitoneal [i.p.]), tiliroside (20 mg/kg,i.p.), sorafenib (30 mg/kg,i.p.), or combination treatment (tiliroside and sorafenib,i.p.). All treatments were administered every 3 d, and the length and width of tumor were measured every 4 d. The formula tumor volume = (length x width2)/2 was used to calculate the tumor volume. Body weight was recorded every 7 d, and the morphology of the tumor was photographed using animal in vivo imaging technology (IVIS Spectrum; PerkinElmer) before the day of sacrifice. The mice were sacrificed 40 d after administration, and the tumors were dissected and weighed. The major organs and xenograft tumors were fixed with 4% paraformaldehyde.
Click to Show/Hide
|
||||
Response regulation | Tiliroside directly binds to TBK1 and inhibits its activity, which inhibits the phosphorylation of Ser349 on p62. Consequently, this decreases the affinity of p62 for Keap1, promotes ubiquitination and degradation of Nrf2 and ferroptosis, and eventually increases the sensitivity of hepatocellular carcinoma cells to sorafenib. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [7] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Tiliroside | Investigative | |||
Responsed Regulator | Kelch-like ECH-associated protein 1 (KEAP1) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
All animal studies were approved by the Committee on Ethics of Animal Experiments of Binzhou Medical University (approval no: BZMU-IACUC-2021-331, date: 09/10/2021). To generate the ectopic HCC mouse models, HepG2-luciferase cells (HepG2 cells transfected with luciferase gene) were suspended in serum-free media and matrigel (BD Biosciences) at a ratio of 1:1 v/v. A total of 2.5 x 106 HepG2-luciferase cells/100 ul were injected into the left axilla of mice. After reaching a tumor size of 100-150 mm3, all mice were randomly divided into four groups: control (vehicle, intraperitoneal [i.p.]), tiliroside (20 mg/kg,i.p.), sorafenib (30 mg/kg,i.p.), or combination treatment (tiliroside and sorafenib,i.p.). All treatments were administered every 3 d, and the length and width of tumor were measured every 4 d. The formula tumor volume = (length x width2)/2 was used to calculate the tumor volume. Body weight was recorded every 7 d, and the morphology of the tumor was photographed using animal in vivo imaging technology (IVIS Spectrum; PerkinElmer) before the day of sacrifice. The mice were sacrificed 40 d after administration, and the tumors were dissected and weighed. The major organs and xenograft tumors were fixed with 4% paraformaldehyde.
Click to Show/Hide
|
||||
Response regulation | Tiliroside directly binds to TBK1 and inhibits its activity, which inhibits the phosphorylation of Ser349 on p62. Consequently, this decreases the affinity of p62 for Keap1, promotes ubiquitination and degradation of Nrf2 and ferroptosis, and eventually increases the sensitivity of hepatocellular carcinoma cells to sorafenib. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Withaferin A | Investigative | |||
Responsed Regulator | Kelch-like ECH-associated protein 1 (KEAP1) | Driver | |||
Pathway Response | Pathways in cancer | hsa05200 | |||
Ferroptosis | hsa04216 | ||||
Cell adhesion molecules | hsa04514 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell invasion | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
SNU-449 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0454 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
Response regulation | Withaferin A may attenuate the metastatic potential and sorafenib resistance by regulating Keap1/Nrf2-associated EMT and ferroptosis. Thus, Withaferin A may serve as a promising agent for Hepatocellular carcinoma therapy, especially for advanced hepatocellular carcinoma. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [3] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocarcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Oxaliplatin | Investigative | |||
Responsed Regulator | LINC01134 (IncRNA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Response regulation | LINC01134 was positively correlated with GPX4 or Nrf2, demonstrating the clinical significance of LINC01134, Nrf2 and GPX4 in OXA resistance of hepatocellular carcinoma. Silenced LINC01134 enhances Oxaliplatin sensitivity by facilitating ferroptosis through GPX4 in hepatocarcinoma. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [9] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Responsed Regulator | Sequestosome-1 (SQSTM1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hepa 1-6 cells | Hepatocellular carcinoma | Mus musculus | CVCL_0327 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
To generate murine subcutaneous tumors, 1 x 106 Hepa16 cells in control shRNA or NRF2 knockdown cells in 200 ul phosphate buffered saline were injected subcutaneously to the right of the dorsal midline in C57BL/6 mice. Once the tumors reached 80-100 mm3 at day seven, mice were randomly allocated into groups and treated with erastin (30 mg/kg intraperitoneal injection [i.p.], twice every other day) and sorafenib (10 mg/kg i.p., once every other day) for two weeks.
Click to Show/Hide
|
||||
Response regulation | Upon exposure to ferroptosis-inducing compounds (e.g., erastin, sorafenib, and buthionine sulfoximine), p62 (SQSTM1) expression prevented NRF2 degradation and enhanced subsequent NRF2 nuclear accumulation through inactivation of Kelch-like ECH-associated protein 1. The status of NRF2 is a key factor that determines the therapeutic response to ferroptosis-targeted therapies in hepatocellular carcinoma cells. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [35] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Camptothecin | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Response regulation | Sorafenib is a potent inducer of ferroptosis used to manage hepatocellular carcinoma (HCC). Nrf2 inhibition by Camptothecin improves sorafenib's sensitivity and reduces sorafenib's resistance via the augmentation of sorafenib's ferroptosis action. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [36] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Sulfhydryl oxidase 1 (QSOX1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | MHCC97-H cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_4972 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
Male 6-week-old BALB/c nude mice were purchased from Charles River Company (Shanghai, China). For subcutaneous mouse model, 5 x106 MHCC97H vector control cells or MHCC97H/QSOX1 cells were implanted into right flanks subcutaneously. At the 7th day following implantation, the mice bear with MHCC97H vector control cells or MHCC97H/QSOX1 cells respectively were randomly separated into two groups: one group were treated with vehicle (0.9% NaCl i.p., once every other day) and another group sorafenib (10 mg/kg i.p., once every other day) for two weeks.
Click to Show/Hide
|
||||
Response regulation | QSOX1 disrupts redox homoeostasis and sensitizes hepatocellular carcinoma cells to oxidative stress by inhibiting activation of the master antioxidant transcription factor NRF2. Mechanistically, QSOX1 restrains EGF-induced EGFR activation by promoting ubiquitination-mediated degradation of EGFR and accelerating its intracellular endosomal trafficking, leading to suppression of NRF2 activity. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [25] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Maleylacetoacetate isomerase (GSTZ1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
SNU-449 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0454 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Mice were divided into five groups as follows: WT + DMSO (control), WT + Sora, Gstz1-/-+DMSO, Gstz1-/-+Sora, and Gstz1-/-+Sora + RSL3. Each group included three male and three female mice. At 2 weeks of age, all mice were administered an intraperitoneal injection of diethylnitrosamine (DEN; Sigma, St. Louis, MO, USA) at a dose of 75 mg/kg. At the third week, the mice were intraperitoneally administered carbon tetrachloride (CCl4; Macklin, Shanghai, China) at 2 ml/kg twice a week for 12 weeks. In the WT + Sora and Gstz1-/-+Sora group, the mice at 22 weeks were administered intraperitoneally sorafenib (30 mg/kg) every 2 days for 4 weeks until euthanasia. In the Gstz1-/-+Sora + RSL3 group, in addition to sorafenib administration as described above, the mice were injected intraperitoneally with RSL3 (10 mg/kg) every 2 days for 4 weeks at the same weeks.
Click to Show/Hide
|
||||
Response regulation | GSTZ1 enhanced sorafenib-induced ferroptosis by inhibiting the NRF2/GPX4 axis in hepatocellular carcinoma (HCC) cells. Combination therapy of sorafenib and GPX4 inhibitor RSL3 may be a promising strategy in HCC treatment. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [37] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform (PIK3CA) | Suppressor | |||
Pathway Response | PI3K-Akt signaling pathway | hsa04151 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Mice weighing between 20 and 23 g were selected and 5 x 106 Hep-G2 cells were subcutaneously injected into their backs. The mice were subsequently divided into the following four groups: control (n = 5), FNDC5 overexpressing (n = 5), FNDC5 overexpressing followed by treatment with the PI3K inhibitor LY294002 (MCE, China), and FNDC5 knockdown (n = 5). Seven days after cell injection, sorafenib (30 mg/kg) was administered to all mice via intraperitoneal injection every alternate day for 4 weeks. The mice in the third group were intraperitoneally injected with LY294002 (25 mg/kg) diluted with DMSO twice a week for 4 weeks.
Click to Show/Hide
|
||||
Response regulation | FNDC5 activated the PI3K/Akt pathway, which in turn promoted the nuclear translocation of Nrf2 and increased the intracellular antioxidant response in Hepatocellular Carcinoma Cells, thereby conferring resistance to ferroptosis. Our study provides novel insights for improving the efficacy of sorafenib. | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [37] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Fibronectin type III domain-containing protein 5 (FNDC5) | Suppressor | |||
Pathway Response | PI3K-Akt signaling pathway | hsa04151 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Mice weighing between 20 and 23 g were selected and 5 x 106 Hep-G2 cells were subcutaneously injected into their backs. The mice were subsequently divided into the following four groups: control (n = 5), FNDC5 overexpressing (n = 5), FNDC5 overexpressing followed by treatment with the PI3K inhibitor LY294002 (MCE, China), and FNDC5 knockdown (n = 5). Seven days after cell injection, sorafenib (30 mg/kg) was administered to all mice via intraperitoneal injection every alternate day for 4 weeks. The mice in the third group were intraperitoneally injected with LY294002 (25 mg/kg) diluted with DMSO twice a week for 4 weeks.
Click to Show/Hide
|
||||
Response regulation | FNDC5 activated the PI3K/Akt pathway, which in turn promoted the nuclear translocation of Nrf2 and increased the intracellular antioxidant response in Hepatocellular Carcinoma Cells, thereby conferring resistance to ferroptosis. Our study provides novel insights for improving the efficacy of sorafenib. | ||||
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target | [38] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Proprotein convertase subtilisin/kexin type 9 (PCSK9) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
HuH-6 cells | Hepatoblastoma | Homo sapiens | CVCL_4381 | ||
In Vivo Model |
Zebrafish were maintained at 28 and in light cycle conditions (12 h). The Casper mutant fish line was purchased from the Zebrafish International Resource Center (ZIRC). For zebrafish xenotransplantation, 48 hours post-fertilization (hpf) zebrafish embryos were dechorionated and anesthetized in egg water solution containing 0.04 mg/mL tricaine (Sigma-Aldrich, St. Louis, MO, USA) before human cell injection. Approximately 200 to 500 fluorescent cells were injected (Eppendorf Femtojet microinjector) into the ducts of Cuvier of each embryo, and the zebrafish were maintained in 0.3X Danieaus solution for 1 h at 28 . After confirmation of a visible cell mass at the injection site, the zebrafish were transferred to a 24-well plate in 500 uL of a 0.3X Danieaus solution incubator and maintained at 34 . The zebrafish with already formed metastasis at 1 hour post-injection (hpi) were discarded.
Click to Show/Hide
|
||||
Response regulation | The disruption of PCSK9 inhibits the anti-ferroptosis p62-Keap1-Nrf2 pathway, one could speculate that a combination therapy of anti-PCSK9 with sorafenib would alleviate drug resistance and improve prognosis. We provide strong evidence supporting the drug repositioning of anti-PCSK9 approaches to treat hepatocellular carcinoma. | ||||
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target | [13] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Long-chain fatty acid transport protein 5 (SLC27A5) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
In Vivo Model |
Age-matched male BALB/c nude mice (4-6 weeks old) were used for the orthotopic mouse model. Cohorts of mice were randomized into different treatment groups. 4 x 106 tumor cells were suspended in a 50 ul PBS/Matrigel (356234, BD Biosciences) mixture (1:1 (v/v) ratio) for each group of mice and injected into the left liver lobes by surgical implantation.
Click to Show/Hide
|
||||
Response regulation | The combination of sorafenib and carmustine (BCNU), a selective inhibitor of GSR, remarkably hamper tumor growth by enhancing ferroptotic cell death in vivo. SLC27A5 serves as a suppressor in sorafenib resistance and promotes sorafenib-triggered ferroptosis via restraining the NRF2/GSR pathway in hepatocellular carcinoma, providing a potential therapeutic strategy for overcoming sorafenib resistance. | ||||
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target | [37] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | RAC-alpha serine/threonine-protein kinase (AKT1) | Suppressor | |||
Pathway Response | PI3K-Akt signaling pathway | hsa04151 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Mice weighing between 20 and 23 g were selected and 5 x 106 Hep-G2 cells were subcutaneously injected into their backs. The mice were subsequently divided into the following four groups: control (n = 5), FNDC5 overexpressing (n = 5), FNDC5 overexpressing followed by treatment with the PI3K inhibitor LY294002 (MCE, China), and FNDC5 knockdown (n = 5). Seven days after cell injection, sorafenib (30 mg/kg) was administered to all mice via intraperitoneal injection every alternate day for 4 weeks. The mice in the third group were intraperitoneally injected with LY294002 (25 mg/kg) diluted with DMSO twice a week for 4 weeks.
Click to Show/Hide
|
||||
Response regulation | FNDC5 activated the PI3K/Akt pathway, which in turn promoted the nuclear translocation of Nrf2 and increased the intracellular antioxidant response in Hepatocellular Carcinoma Cells, thereby conferring resistance to ferroptosis. Our study provides novel insights for improving the efficacy of sorafenib. | ||||
Long-chain-fatty-acid--CoA ligase 4 (ACSL4)
In total 5 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Lactate | Investigative | |||
Responsed Regulator | Hydroxycarboxylic acid receptor 1 (HCAR1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
AMPK signaling pathway | hsa04152 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | CAF cells | Normal | Carassius auratus | CVCL_R883 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Female mice aged around 6-7 weeks were used for this study, which were purchased through Laboratory Animal Center of Chongqing Medical University from Vital River Co. Ltd (Beijing, China).After one week, each mouse was injected subcutaneously with 100 uL of Huh-7 cell suspension (5 x 106 units) to establish the tumor model. The mice were grouped randomly, and then subjected to different treatments after subcutaneous tumors became visually detectable.
Click to Show/Hide
|
||||
Response regulation | Lactate regulates the ferroptosis of hepatocellular carcinoma cells. And blocking the lactate uptake via hydroxycarboxylic acid receptor 1 (HCAR1)/MCT1 inhibition promotes ferroptosis by activating the AMPK to downregulate SCD1, which may synergize with its acyl-coenzyme A synthetase 4 (ACSL4)-promoting effect to amplify the ferroptotic susceptibility. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [10] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Responsed Regulator | Protein C-ets-1 (ETS1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | MHCC97-L cells | Hepatocellular carcinoma | Homo sapiens | CVCL_4973 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
HEK293 FT cells | Normal | Homo sapiens | CVCL_6911 | ||
In Vivo Model |
Parental MHCC97L cells (2 x 106 cells/mouse) were subcutaneously injected into the 4-to-5-week-old NOD-SCID mice. When the tumours reached a volume of around 50-100 mm3 (calculated by the formula 4/3(D/2)(d/2)2, where D and d represent the minor and major axis of the tumour, respectively), the maximum tolerated dose of sorafenib (50 mg/kg) was given to the mice by oral gavage daily until the drug resistance occurred, denoted as the drug resistant group. For the control, the wild type group was treated with the vehicle (0.5% CMC-Na). The tumour size and body weight were measured every 3 days.
Click to Show/Hide
|
||||
Response regulation | Sorafenib treatment triggered ferroptosis via lipid ROS production and chelatable iron accumulation. The ETS1 upregulated by sorafenib was a key transcription factor of miR-23a-3p that directly enhanced miR-23a-3p expression. MiR-23a-3p recognized and bound to ACSL4 3UTR to limit lipid ROS production, thus attenuating sorafenib-induced ferroptotic cell death in hepatocellular carcinoma. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [10] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Responsed Regulator | hsa-miR-23a-3p (miRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | MHCC97-L cells | Hepatocellular carcinoma | Homo sapiens | CVCL_4973 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
HEK293 FT cells | Normal | Homo sapiens | CVCL_6911 | ||
In Vivo Model |
Parental MHCC97L cells (2 x 106 cells/mouse) were subcutaneously injected into the 4-to-5-week-old NOD-SCID mice. When the tumours reached a volume of around 50-100 mm3 (calculated by the formula 4/3(D/2)(d/2)2, where D and d represent the minor and major axis of the tumour, respectively), the maximum tolerated dose of sorafenib (50 mg/kg) was given to the mice by oral gavage daily until the drug resistance occurred, denoted as the drug resistant group. For the control, the wild type group was treated with the vehicle (0.5% CMC-Na). The tumour size and body weight were measured every 3 days.
Click to Show/Hide
|
||||
Response regulation | Sorafenib treatment triggered ferroptosis via lipid ROS production and chelatable iron accumulation. The ETS1 upregulated by sorafenib was a key transcription factor of miR-23a-3p that directly enhanced miR-23a-3p expression. MiR-23a-3p recognized and bound to ACSL4 3UTR to limit lipid ROS production, thus attenuating sorafenib-induced ferroptotic cell death in hepatocellular carcinoma. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [18] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Atractylodin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hccm (Human hepatocellular carcinoma cells) | |||||
Response regulation | Atractylodin can inhibit the proliferation, migration, and invasion of Huh7 and Hccm liver cancer cells, and induce cell apoptosis and cell cycle arrest. In addition, atractylodin may induce ferroptosis in hepatocellular carcinoma cells by inhibiting the expression of GPX4 and FTL proteins, and up-regulating the expression of ACSL4 and TFR1 proteins. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [23] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Seco-Lupane Triterpene Derivatives | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | A new seco-lupane triterpene derivative, compound21, was found to regulate cell growth through the cell cycle and ferroptosis, which in turn inhibited the proliferation, migration, and invasion of HepG2 cells. And it was found that compound 21 significantly upregulated ACSL4 protein expression and downregulated GPX4 protein expression. It has the potential to become an effective new drug for the treatment of hepatocellular carcinoma. | ||||
Unspecific Target
In total 35 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [11] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | RSL3 | Investigative | |||
Responsed Regulator | Ceruloplasmin (CP) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
Response regulation | Erastin and RSL3 suppress ceruloplasmin expression in hepatocellular carcinoma cells. CP suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma cells. The suppression function of ceruloplasmin in erastin- and RSL3-induced ferroptosis is dependent on FPN. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [12] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Cathepsin B (CTSB) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
SNU-182 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0090 | ||
SNU-449 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0454 | ||
In Vivo Model |
A total of 20 male Balb/c nude mice aged 6-8 weeks were purchased from Hunan SJA Laboratory Animal Co., Ltd. (Changsha, China). Five million Huh7 cells were inoculated into the right flanks of the mice. When the tumor size reached 80-100 mm3, the mice were randomly divided into four groups and administered artesunate (30 mg/kg mouse weight) alone, sorafenib (20 mg/kg mouse weight) alone, a combination of artesunate and sorafenib, or the same volume of PBS by gavage every other day.
Click to Show/Hide
|
||||
Response regulation | Sorafenib at low dose mainly caused oxidative stress through mitochondrial impairments and SLC7A11-invovled glutathione depletion. Artesunate-induced lysosome activation synergized with sorafenib-mediated pro-oxidative effects by promoting sequential reactions including lysosomal cathepsin B/L activation, ferritin degradation, lipid peroxidation, and consequent ferroptosis. Taken together, artesunate could be repurposed to sensitize sorafenib in hepatocellular carcinoma treatment. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [13] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Carmustine | Investigative | |||
Responsed Regulator | Glutathione reductase, mitochondrial (GSR) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
In Vivo Model |
Age-matched male BALB/c nude mice (4-6 weeks old) were used for the orthotopic mouse model. Cohorts of mice were randomized into different treatment groups. 4 x 106 tumor cells were suspended in a 50 ul PBS/Matrigel (356234, BD Biosciences) mixture (1:1 (v/v) ratio) for each group of mice and injected into the left liver lobes by surgical implantation.
Click to Show/Hide
|
||||
Response regulation | The combination of sorafenib and carmustine (BCNU), a selective inhibitor of GSR, remarkably hamper tumor growth by enhancing ferroptotic cell death in vivo. SLC27A5 serves as a suppressor in sorafenib resistance and promotes sorafenib-triggered ferroptosis via restraining the NRF2/GSR pathway in hepatocellular carcinoma, providing a potential therapeutic strategy for overcoming sorafenib resistance. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [11] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Erastin | Investigative | |||
Responsed Regulator | Ceruloplasmin (CP) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
Response regulation | Erastin and RSL3 suppress ceruloplasmin expression in hepatocellular carcinoma cells. CP suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma cells. The suppression function of ceruloplasmin in erastin- and RSL3-induced ferroptosis is dependent on FPN. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [14] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Erastin | Investigative | |||
Responsed Regulator | GA-binding protein subunit beta-1 (GABPB1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Response regulation | Erastin upregulated the lncRNA GABPB1-AS1, which downregulated GABPB1 protein levels by blocking GABPB1 translation, leading to the downregulation of the gene encoding Peroxiredoxin-5 (PRDX5) peroxidase and the eventual suppression of the cellular antioxidant capacity. GABPB1 and GABPB1-AS1 are attractive therapeutic targets for hepatocellular carcinoma. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [14] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Erastin | Investigative | |||
Responsed Regulator | Peroxiredoxin-5, mitochondrial (PRDX5) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Response regulation | Erastin upregulated the lncRNA GABPB1-AS1, which downregulated GABPB1 protein levels by blocking GABPB1 translation, leading to the downregulation of the gene encoding Peroxiredoxin-5 (PRDX5) peroxidase and the eventual suppression of the cellular antioxidant capacity. GABPB1 and GABPB1-AS1 are attractive therapeutic targets for hepatocellular carcinoma. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [14] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Erastin | Investigative | |||
Responsed Regulator | GABPB1-AS1 (IncRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Response regulation | Erastin upregulated the lncRNA GABPB1-AS1, which downregulated GABPB1 protein levels by blocking GABPB1 translation, leading to the downregulation of the gene encoding Peroxiredoxin-5 (PRDX5) peroxidase and the eventual suppression of the cellular antioxidant capacity. GABPB1 and GABPB1-AS1 are attractive therapeutic targets for hepatocellular carcinoma. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [15] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Heteronemin | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell apoptosis | |||||
In Vitro Model | HA22T/VGH cells | Hepatocellular carcinoma | Homo sapiens | CVCL_7046 | |
HA59T/VGH cells | Hepatocellular carcinoma | Homo sapiens | CVCL_Y018 | ||
Response regulation | Heteronemin is an effective agent against hepatocellular carcinoma that induces HCC cell apoptosis and ferroptosis by inducing intracellular ROS formation and the p38 (MAPK14)/JNK MAPK signaling pathway, revealing the potent MAPK-mediated crosstalk mechanism between apoptosis and ferroptosis. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [16] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Responsed Regulator | Ragulator complex protein LAMTOR5 (LAMTOR5) | Suppressor | |||
Pathway Response | Glutathione metabolism | hsa00480 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
In Vivo Model |
Six-week-old male BALB/c athymic nude mice were purchased from the Experimental Animal Center of Peking (Beijing, China). Stable cells (5 x 106) were seeded into the right flanks of the mice. After the xenografts had grown to 200 mm3, saline as a vehicle or sorafenib (30 mg/kg) was administered by gavage every day, and the mice were euthanized by the cervical dislocation method five weeks later. Before sacrifice, the tumor sizes and body weights were measured twice per week. The tumor volume (V) was calculated as follows: (L x W2)/2 (length, L, and width, W). The xenografts were excised and further assessed.
Click to Show/Hide
|
||||
Response regulation | Sorafenib decreased HBXIP expression, and overexpression of HBXIP blocked sorafenib-induced hepatocellular carcinoma cell death. Regarding the molecular mechanism, HBXIP transcriptionally induced the expression of stearoyl-CoA desaturase (SCD) via coactivating the transcriptional factor ZNF263, resulting in the accumulation of free fatty acids and suppression of ferroptosis. | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [17] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | ZZW-115 | Investigative | |||
Responsed Regulator | Transcription factor A, mitochondrial (TFAM) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | MIA PaCa-2 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0428 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
In Vivo Model |
Xenografts with MiaPaCa-2 and HepG2 cells in nude mice and treated them for 4 or 3 weeks, respectively, with vehicle alone and 2.5 or 5.0 mg/kg/day of ZZW-115. Then, we measured the GPX4 activity and analyzed the mRNA levels of the key genes involved in ferroptosis by qRT-PCR analysis.
Click to Show/Hide
|
||||
Response regulation | The expression of TFAM, a key regulator of mitochondrial biogenesis, is downregulated by ZZW-115. Forced expression of TFAM is able to rescue morphological and functional mitochondrial alterations, ROS production, and cell death induced by ZZW-115 or genetic inhibition of NUPR1. These results have been validated in xenografts induced with pancreatic ductal adenocarcinoma (PDAC)- and hepatocellular carcinoma (HCC)-derived cells in nude mice during the treatment with ZZW-115. | ||||
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target | [17] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | ZZW-115 | Investigative | |||
Responsed Regulator | Nuclear protein 1 (NUPR1) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | MIA PaCa-2 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0428 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
In Vivo Model |
Xenografts with MiaPaCa-2 and HepG2 cells in nude mice and treated them for 4 or 3 weeks, respectively, with vehicle alone and 2.5 or 5.0 mg/kg/day of ZZW-115. Then, we measured the GPX4 activity and analyzed the mRNA levels of the key genes involved in ferroptosis by qRT-PCR analysis.
Click to Show/Hide
|
||||
Response regulation | The expression of TFAM, a key regulator of mitochondrial biogenesis, is downregulated by ZZW-115. Forced expression of TFAM is able to rescue morphological and functional mitochondrial alterations, ROS production, and cell death induced by ZZW-115 or genetic inhibition of NUPR1. These results have been validated in xenografts induced with pancreatic ductal adenocarcinoma (PDAC)- and hepatocellular carcinoma (HCC)-derived cells in nude mice during the treatment with ZZW-115. | ||||
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target | [53] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | 1-Methylnaphthalene-4-propionate endoperoxide | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hepa 1-6 cells | Hepatocellular carcinoma | Mus musculus | CVCL_0327 | |
Response regulation | 1-Methylnaphthalene-4-propionate endoperoxide (MNPE) is a cell-permeable endoperoxide that generates pure singlet oxygen. MNPE treatment decreased the viabilities of mouse liver cancer cells in vitro, and that this decrease was accompanied by increases in the concentrations of both intracellular ferrous iron and the level of lipid peroxidation. | ||||
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target | [54] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Boric Acid | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | High boric acid (BA) concentrations can directly induce ferroptosis in hepatocellular carcinoma, and such BA-induced ferroptosis can add to the cytotoxic effects of anticancer drugs sorafenib, doxorubicin and cisplatin. | ||||
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target | [55] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Aldo-keto reductase family 1 member C3 (AKR1C3) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
PI3K-Akt signaling pathway | hsa04151 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Response regulation | AKR1C3 can induce sorafenib resistance through promoting the phosphorylation of AKT in hepatocellular carcinoma (HCC). AKR1C3 inhibitors may be used in conjunction with sorafenib to become a better therapeutic target for HCC. | ||||
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target | [56] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | cIARS (circRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Response regulation | cIARS (hsa_circ_0008367) was found to be the most highly expressed circRNA after sorafenib (SF) treatment in hepatocellular carcinoma cells. cIARS may be an important circRNA, positively regulating SF-induced ferroptosis through suppressing the ALKBH5-mediated autophagy inhibition. | ||||
Experiment 16 Reporting the Ferroptosis-centered Disease Response by This Target | [57] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Circ_CMTM3 (circRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
HCCLM3 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_6832 | ||
THLE-2 cells | Normal | Homo sapiens | CVCL_3803 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
In Vivo Model |
Six-week-old BALB/c female athymic nude mice (SJA Laboratory Animal Co., Ltd, China) were used to construct the xenograft. The mice were randomly grouped: (1) shNC + vehicle; (2) shNC + Erastin; (3) shWTAP + Erastin; (4) shcircCMTM3 + Erastin. Briefly, HCC cells with stable silenced WTAP and circCMTM3 (2 x 106 in 0.1 mL PBS) were injected into the left flank of the mice subcutaneously. Once the tumor size reached approximately > 60 mm3, mice in the groups (2), (3), and (4) were injected with 15 mg/kg erastin. Erastin was injected twice a day for a period of time.
Click to Show/Hide
|
||||
Response regulation | CircCMTM3 promoted the carcinogenesis through inhibiting ferroptosis by recruiting IGF2BP1 to increase PARK7 stability in hepatocellular carcinoma (HCC), suggesting that cicrCMTM3 may be an important marker for HCC treatment. | ||||
Experiment 17 Reporting the Ferroptosis-centered Disease Response by This Target | [58] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Collectrin (CLTRN) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | CLTRN acts as a target of radiation, which is regulated by the NRF1/RAN/DLD protein complex, and it enhances the radiosensitivity of hepatocellular carcinoma cells through ferroptosis. | ||||
Experiment 18 Reporting the Ferroptosis-centered Disease Response by This Target | [59] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Gamma-aminobutyric acid receptor-associated protein-like 1 (GABARAPL1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | SNU-449 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0454 | |
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
MHCC97-H cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_4972 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
LM3 cells | Malignant neoplasms | Mus musculus | CVCL_D269 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
Response regulation | GABARAPL1 was downregulated in hepatocellular carcinoma (HCC) tumor-repopulating cells (TRC; a type of CSLC). Its downregulation decreased the sensitivity of HCC TRC to erastin- or sorafenib-triggered ferroptosis. | ||||
Experiment 19 Reporting the Ferroptosis-centered Disease Response by This Target | [60] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Leukemia inhibitory factor receptor (LIFR) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Mahlavu cells | Hepatoma | Homo sapiens | CVCL_0405 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
HA22T/VGH cells | Hepatocellular carcinoma | Homo sapiens | CVCL_7046 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
PDX tumors in cold Dulbeccos Modified Eagles Medium (DMEM) were minced into 1-2 mm3 fragments, and each fragment was subcutaneously transplanted into the dorsal flank of 6-week-old male NSG (non-obese diabetic; severe combined immunodeficiency; interleukin-2 receptor gamma chain null) mice. Tumor growth was monitored by bidimensional tumor measurements with a caliper twice a week until the endpoint.
Click to Show/Hide
|
||||
Response regulation | LIFR and SHP1 (PTPN6) positively regulate ferroptosis while LCN2 negatively regulates ferroptosis. Notably, an LCN2-neutralizing antibody enhances the ferroptosis-inducing and anticancer effects of sorafenib on hepatocellular carcinoma patient-derived xenograft tumors with low LIFR expression and high LCN2 expression. | ||||
Experiment 20 Reporting the Ferroptosis-centered Disease Response by This Target | [61] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Metallothionein-1G (MT1G) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
To generate murine subcutaneous tumors, 2 x 106 Huh7 cells in control shRNA or MT-1G knockdown cells in 200 ul phosphate buffered saline were injected subcutaneously to the right of the dorsal midline in nude mice. Once the tumors reached 80-100 mm3 at day seven, mice were randomly allocated into groups and treated with sorafenib (10 mg/kg/intraperitoneal injection (i.p.), once every other day) for two weeks. In another experiment, nude mice were injected subcutaneously with indicated Huh7 cells (2 x 106 cells/mouse) and treated with sorafenib (10 mg/kg/i.p., once every other day) with or without ATRA (0.5 mg/kg/i.p., once every other day) or PPG (10 mg/kg/i.p., once every other day) at day seven for two weeks.
Click to Show/Hide
|
||||
Response regulation | MT-1G is a critical regulator and promising therapeutic target of sorafenib resistance in human hepatocellular carcinoma cells. Knockdown of MT-1G by RNA interference increases glutathione depletion and lipid peroxidation, which contributes to sorafenib-induced ferroptosis. | ||||
Experiment 21 Reporting the Ferroptosis-centered Disease Response by This Target | [62] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | NEAT1 (IncRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
NU/NU nude mice were purchased from Charles River (Beijing). For xenograft models, HepG2 or HuH-7 cells (5 x 106 cells per mouse) that were transfected with the NEAT1 vector or an empty vector were injected into the left posterior flanks of 7-week-old immunodeficient female nude mice. The tumors were measured every 4 days, and tumor volume was calculated using the following formula: volume = (L x W2)/2, among which L and W are the longest and shortest diameters, respectively. When tumors reached a volume of ~50 mm3, mice were randomly allocated into groups and treated with erastin or RSL3 via intraperitoneal injection for 20 days. Mice were then sacrificed, the xenograft tumors were excised and weighted for immunohistochemistry assays. The erastin was dissolved in 5% DMSO + corn oil (C8267, Sigma). To better dissolve erastin, we warmed the tube at 37 water base and shake it gently.
Click to Show/Hide
|
||||
Response regulation | NEAT1 can competitively bind more miR-362-3p and thus leads to less miR-362-3p-mediated MIOX inhibition, thereby increasing the sensitivity of hepatocellular carcinoma cells to ferroptosis. | ||||
Experiment 22 Reporting the Ferroptosis-centered Disease Response by This Target | [63] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | Ribonucleoside-diphosphate reductase subunit M2 (RRM2) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | |
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
BEL-7404 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6568 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Response regulation | RRM2 exerts an anti-ferroptotic role in liver cancer cells by sustaining GSH synthesis. Serum RRM2 will be useful as a biomarker to evaluate the degree to which ferroptosis is suppressed and improve diagnostic efficiency for liver cancer. | ||||
Experiment 23 Reporting the Ferroptosis-centered Disease Response by This Target | [64] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | Transcription factor Jun (JUN) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | |
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
In Vivo Model |
Eight-week-old athymic nude mice were obtained from Bikai Laboratory Animal Crop (Bikai, Shanghai, China) and Bel-7402 cells (initial 5 x 106) were subcutaneously injected into each mouse. Dimethy sulfoxide (DMSO) or piperazine erastin (5 mg/kg, MedChemExpress, Monmouth Junction, NJ, USA) was subcutaneously injected once every day after xenografts were formed.
Click to Show/Hide
|
||||
Response regulation | A positive correlation between c-Jun O-GlcNAcylation and GSH was observed in clinical samples. Collectively, O-GlcNAcylated c-Jun represents an obstructive factor to ferroptosis, and targeting O-GlcNAcylated c-Jun might be helpful for treating liver cancer. | ||||
Experiment 24 Reporting the Ferroptosis-centered Disease Response by This Target | [65] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Cystathionine beta-synthase (CBS) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell cycle | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
MDA-MB-231 cells | Breast adenocarcinoma | Homo sapiens | CVCL_0062 | ||
PANC-28 cells | Pancreatic adenocarcinoma | Homo sapiens | CVCL_3917 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
mEFs (Mouse embryonic fibroblasts) | |||||
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | ||
H22 cells | Hepatoma | Mus musculus | CVCL_H613 | ||
In Vivo Model |
H22 mouse liver tumor cells were intraperitoneal injected into female ICR mice (SLAC Laboratory Animal Co. Ltd, Shanghai, China). 8 days later, mice with distended abdomen were killed, and the ascites was collected, washed and resuspended with PBS. 0.1ml cell suspension with a density of 1 x 107/ml was subcutaneously injected into the right dorsum of ICR female mice (~20g). The mice bearing H22 tumor cells were then randomized into groups (n = 8) on the following day after the implantation. Then, the mice were injected via a tail vein (i.v.) with PBS, CH004 (10 mg/kg) or cyclophosphamide (CTX, a known anticancer drug; 20 mg/kg) once per day and for 21 days.
Click to Show/Hide
|
||||
Response regulation | Chemical or genetic inhibition of CBS demonstrates that endogenous CBS is closely coupled with cell proliferation and cell cycle. Moreover, CH004 substantially retarded in vivo tumor growth in a xenograft mice model of liver cancer. Importantly, inhibition of CBS triggers ferroptosis in hepatocellular carcinoma. | ||||
Experiment 25 Reporting the Ferroptosis-centered Disease Response by This Target | [56] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | RNA demethylase ALKBH5 (ALKBH5) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Response regulation | cIARS (hsa_circ_0008367) was found to be the most highly expressed circRNA after sorafenib (SF) treatment in hepatocellular carcinoma cells. cIARS may be an important circRNA, positively regulating SF-induced ferroptosis through suppressing the ALKBH5-mediated autophagy inhibition. | ||||
Experiment 26 Reporting the Ferroptosis-centered Disease Response by This Target | [57] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Parkinson disease protein 7 (PARK7) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
HCCLM3 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_6832 | ||
THLE-2 cells | Normal | Homo sapiens | CVCL_3803 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
In Vivo Model |
Six-week-old BALB/c female athymic nude mice (SJA Laboratory Animal Co., Ltd, China) were used to construct the xenograft. The mice were randomly grouped: (1) shNC + vehicle; (2) shNC + Erastin; (3) shWTAP + Erastin; (4) shcircCMTM3 + Erastin. Briefly, HCC cells with stable silenced WTAP and circCMTM3 (2 x 106 in 0.1 mL PBS) were injected into the left flank of the mice subcutaneously. Once the tumor size reached approximately > 60 mm3, mice in the groups (2), (3), and (4) were injected with 15 mg/kg erastin. Erastin was injected twice a day for a period of time.
Click to Show/Hide
|
||||
Response regulation | CircCMTM3 promoted the carcinogenesis through inhibiting ferroptosis by recruiting IGF2BP1 to increase PARK7 stability in hepatocellular carcinoma (HCC), suggesting that cicrCMTM3 may be an important marker for HCC treatment. | ||||
Experiment 27 Reporting the Ferroptosis-centered Disease Response by This Target | [60] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Tyrosine-protein phosphatase non-receptor type 6 (PTPN6) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Mahlavu cells | Hepatoma | Homo sapiens | CVCL_0405 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
HA22T/VGH cells | Hepatocellular carcinoma | Homo sapiens | CVCL_7046 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
PDX tumors in cold Dulbeccos Modified Eagles Medium (DMEM) were minced into 1-2 mm3 fragments, and each fragment was subcutaneously transplanted into the dorsal flank of 6-week-old male NSG (non-obese diabetic; severe combined immunodeficiency; interleukin-2 receptor gamma chain null) mice. Tumor growth was monitored by bidimensional tumor measurements with a caliper twice a week until the endpoint.
Click to Show/Hide
|
||||
Response regulation | LIFR and SHP1 (PTPN6) positively regulate ferroptosis while LCN2 negatively regulates ferroptosis. Notably, an LCN2-neutralizing antibody enhances the ferroptosis-inducing and anticancer effects of sorafenib on hepatocellular carcinoma patient-derived xenograft tumors with low LIFR expression and high LCN2 expression. | ||||
Experiment 28 Reporting the Ferroptosis-centered Disease Response by This Target | [60] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Neutrophil gelatinase-associated lipocalin (LCN2) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Mahlavu cells | Hepatoma | Homo sapiens | CVCL_0405 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
HA22T/VGH cells | Hepatocellular carcinoma | Homo sapiens | CVCL_7046 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
PDX tumors in cold Dulbeccos Modified Eagles Medium (DMEM) were minced into 1-2 mm3 fragments, and each fragment was subcutaneously transplanted into the dorsal flank of 6-week-old male NSG (non-obese diabetic; severe combined immunodeficiency; interleukin-2 receptor gamma chain null) mice. Tumor growth was monitored by bidimensional tumor measurements with a caliper twice a week until the endpoint.
Click to Show/Hide
|
||||
Response regulation | LIFR and SHP1 (PTPN6) positively regulate ferroptosis while LCN2 negatively regulates ferroptosis. Notably, an LCN2 -neutralizing antibody enhances the ferroptosis-inducing and anticancer effects of sorafenib on hepatocellular carcinoma patient-derived xenograft tumors with low LIFR expression and high LCN2 expression. | ||||
Experiment 29 Reporting the Ferroptosis-centered Disease Response by This Target | [62] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | hsa-miR-362-3p (miRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
NU/NU nude mice were purchased from Charles River (Beijing). For xenograft models, HepG2 or HuH-7 cells (5 x 106 cells per mouse) that were transfected with the NEAT1 vector or an empty vector were injected into the left posterior flanks of 7-week-old immunodeficient female nude mice. The tumors were measured every 4 days, and tumor volume was calculated using the following formula: volume = (L x W2)/2, among which L and W are the longest and shortest diameters, respectively. When tumors reached a volume of ~50 mm3, mice were randomly allocated into groups and treated with erastin or RSL3 via intraperitoneal injection for 20 days. Mice were then sacrificed, the xenograft tumors were excised and weighted for immunohistochemistry assays. The erastin was dissolved in 5% DMSO + corn oil (C8267, Sigma). To better dissolve erastin, we warmed the tube at 37 water base and shake it gently.
Click to Show/Hide
|
||||
Response regulation | NEAT1 can competitively bind more miR-362-3p and thus leads to less miR-362-3p-mediated MIOX inhibition, thereby increasing the sensitivity of hepatocellular carcinoma cells to ferroptosis. | ||||
Experiment 30 Reporting the Ferroptosis-centered Disease Response by This Target | [62] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Inositol oxygenase (MIOX) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
NU/NU nude mice were purchased from Charles River (Beijing). For xenograft models, HepG2 or HuH-7 cells (5 x 106 cells per mouse) that were transfected with the NEAT1 vector or an empty vector were injected into the left posterior flanks of 7-week-old immunodeficient female nude mice. The tumors were measured every 4 days, and tumor volume was calculated using the following formula: volume = (L x W2)/2, among which L and W are the longest and shortest diameters, respectively. When tumors reached a volume of ~50 mm3, mice were randomly allocated into groups and treated with erastin or RSL3 via intraperitoneal injection for 20 days. Mice were then sacrificed, the xenograft tumors were excised and weighted for immunohistochemistry assays. The erastin was dissolved in 5% DMSO + corn oil (C8267, Sigma). To better dissolve erastin, we warmed the tube at 37 water base and shake it gently.
Click to Show/Hide
|
||||
Response regulation | NEAT1 can competitively bind more miR-362-3p and thus leads to less miR-362-3p-mediated MIOX inhibition, thereby increasing the sensitivity of hepatocellular carcinoma cells to ferroptosis. | ||||
Experiment 31 Reporting the Ferroptosis-centered Disease Response by This Target | [66] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | RB1-inducible coiled-coil protein 1 (RB1CC1) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
MKN45 cells | Gastric adenocarcinoma | Homo sapiens | CVCL_0434 | ||
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
PC-9 cells | Lung adenocarcinoma | Homo sapiens | CVCL_B260 | ||
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
NCI-H226 cells | Pleural epithelioid mesothelioma | Homo sapiens | CVCL_1544 | ||
SW1990 cells | Pancreatic adenocarcinoma | Homo sapiens | CVCL_1723 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
HT-29 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0320 | ||
NCI-H460 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0459 | ||
Calu-1 cells | Lung squamous cell carcinoma | Homo sapiens | CVCL_0608 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
143B cells | Osteosarcoma | Homo sapiens | CVCL_2270 | ||
In Vivo Model |
To evaluate the effects of drugs to strengthen the effects of IKE, cell-derived xenograft (CDX) mouse models were generated by subcutaneously injecting 2 x 106 cells per athymic nude mouse (BALB/c-nu, Spaefer, Beijing, China). When tumours were 220-250 mm3, mice were administered with IKE (50 mg/kg) daily with or without PTX (20 mg/kg), Clolar (10 mg/kg), OXA (10 mg/kg) or TMZ (40 mg/kg). Tumour growth was monitored, and sizes were calculated by 0.5 x L x W2 (L indicating length and W indicating width).
Click to Show/Hide
|
||||
Response regulation | Nuclear localisation of RB1CC1 correlated with lipid peroxidation in clinical lung cancer specimens. Rb1cc1 was essential for ferroptosis agonists to suppress liver tumourigenesis in mice. | ||||
Experiment 32 Reporting the Ferroptosis-centered Disease Response by This Target | [44] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Cellular tumor antigen p53 (TP53) | Driver | |||
Pathway Response | Autophagy | hsa04140 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
In Vivo Model |
Xenograft mouse model experiments were used male BALB/c nude mice (4 weeks old) purchased from SPF Biotechnology (Beijing, China). Each mouse was injected 5 x 106 tumor cells at the volume of 100 uL into the subcutaneous tissue. The tumor volume and weight of the mice was observed every 2 days. Mice were monitored daily and the tumor volume calculated according to the equation volume = length x width2 x 1/2.
Click to Show/Hide
|
||||
Response regulation | PNO1 inhibits autophagy-mediated ferroptosis via GSH metabolic reprogramming as demonstrated above. We also demonstrated that PNO1 inhibition repressed SLC7A11 through p53 to promote ferroptosis. These observations suggested that sh-PNO1 could be a new target in hepatocellular carcinoma therapy. | ||||
Experiment 33 Reporting the Ferroptosis-centered Disease Response by This Target | [67] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | hsa-mir-214 (Precursor RNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
The 5-week-old nude mice (BALB/c) were purchased from Beijing HFK Bioscience Co., Ltd. (China). Hep3B cells (7 x 106) stably transfected with NC1 or pre-miR-214 plasmid were subcutaneously injected into the nude mice. When the tumor sizes reached approximately >50 mm3, mice in Groups B and C were treated with 15 mg/kg erastin (MCE, China) twice every day for 20 days. Mice in Group A were treated with an equal volume vehicle.
Click to Show/Hide
|
||||
Response regulation | The ferroptosis-promoting effects of miR-214 in hepatocellular carcinoma cells are attributed at least to its inhibitory effects on ATF4, which may provide a new target for therapy of hepatoma regarding ferroptosis. | ||||
Experiment 34 Reporting the Ferroptosis-centered Disease Response by This Target | [67] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Cyclic AMP-dependent transcription factor ATF-4 (ATF4) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
The 5-week-old nude mice (BALB/c) were purchased from Beijing HFK Bioscience Co., Ltd. (China). Hep3B cells (7 x 106) stably transfected with NC1 or pre-miR-214 plasmid were subcutaneously injected into the nude mice. When the tumor sizes reached approximately >50 mm3, mice in Groups B and C were treated with 15 mg/kg erastin (MCE, China) twice every day for 20 days. Mice in Group A were treated with an equal volume vehicle.
Click to Show/Hide
|
||||
Response regulation | The ferroptosis-promoting effects of miR-214 in hepatocellular carcinoma cells are attributed at least to its inhibitory effects on ATF4, which may provide a new target for therapy of hepatoma regarding ferroptosis. | ||||
Experiment 35 Reporting the Ferroptosis-centered Disease Response by This Target | [68] | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Retinoblastoma-associated protein (RB1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_6714 | ||
In Vivo Model |
Six week-old female Balb/cnude mice (Janvier, Le Genest Saint Isle, France) were injected with HCC cells (10 x 106 cells) stably transfected with the vector pGL4.51 [luc2/CMV/neo] (Promega) encoding the luciferase reporter gene luc2 (Photinus pyralis). When the first tumours appeared, mice were randomly assigned into four groups: control or shRb, receiving sorafenib administered by oral gavage (30 mg/kg/d, resuspended in Cremophore EL) or vehicle alone.
Click to Show/Hide
|
||||
Response regulation | Upon exposure to sorafenib, the Rb (RB1)-negative status of hepatocellular carcinoma cells promotes the occurrence of ferroptosis, a form of oxidative necrosis. The findings highlight the role of Rb in the response of HCC cells to sorafenib and the regulation of ferroptosis. | ||||
Transferrin receptor protein 1 (TFRC)
In total 2 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [18] | |||
Target for Ferroptosis | Marker/Suppressor/Driver | |||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | |||
Responsed Drug | Atractylodin | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Apoptosis | hsa04210 | |||
Cell Process | Cell ferroptosis | |||
Cell apoptosis | ||||
Cell proliferation | ||||
Cell migration | ||||
Cell invasion | ||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 |
Hccm (Human hepatocellular carcinoma cells) | ||||
Response regulation | Atractylodin can inhibit the proliferation, migration, and invasion of Huh7 and Hccm liver cancer cells, and induce cell apoptosis and cell cycle arrest. In addition, atractylodin may induce ferroptosis in hepatocellular carcinoma cells by inhibiting the expression of GPX4 and FTL proteins, and up-regulating the expression of ACSL4 and TFR1 proteins. | |||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | |||
Target for Ferroptosis | Marker/Suppressor/Driver | |||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | |||
Responsed Regulator | Tribbles homolog 2 (TRIB2) | Suppressor | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Ubiquitin mediated proteolysis | hsa04120 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model | BEL-7404 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6568 |
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | |
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
Response regulation | The effects by which TrCP-mediated ubiquitination and followed degradation of TFRC to decline labile iron are critical for TRIB2 to desensitize liver cancer cells to ferroptosis. Appropriate reduction of TRIB2 function might be beneficial for patients bearing liver cancer because it will definitely sensitize ferroptosis-based therapy. | |||
Serotransferrin (TF)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [20] | ||||
Target for Ferroptosis | Marker/Suppressor/Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | Peroxisome proliferator-activated receptor alpha (PPARA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hepa 1-6 cells | Hepatocellular carcinoma | Mus musculus | CVCL_0327 | |
In Vivo Model |
C57BL/6J SPF mice were purchased from Huazhong Agricultural University Experimental Animal Center. Mice were given tertian intraperitoneal injections of either PBS (control) or dextriferron (500 mg/kg body weight) for 2 weeks and then sacrificed. Mice were given a daily intraperitoneal injection of either vehicle or ferrostatin-1 (Fer1, 1 mg/kg body weight) for 3 weeks before sacrificed.
Click to Show/Hide
|
||||
Response regulation | PPARa activation alleviates iron overload-induced ferroptosis in mouse livers through Gpx4 and TRF (TF), suggesting that PPAR may be a promising therapeutic target for drug discovery in ferroptosis-related tissue injuries in Hepatocellular carcinomaHepatocellular carcinoma. | ||||
Nuclear receptor coactivator 4 (NCOA4)
In total 4 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [21] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | 2-pyridylhydrazone dithiocarbamate s-acetic acid | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
Cell apoptosis | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | 2-pyridylhydrazone dithiocarbamate s-acetic acid (PdtaA) induced both apoptosis and cell cycle arrest. Notably, PdtaA also induced ferroptosis via downregulation of GPx4 and xCT in liver cancer cells. Autophagy inhibitor 3-methyladenin or genetic knockdown of NCOA4 was employed to inhibit ferritinophagy, which significantly neutralized the action of PdtaA in both apoptosis and ferroptosis. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [32] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | 2-pyridylhydrazone dithiocarbamate s-acetic acid | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell adhesion molecules | hsa04514 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | Knockdown of NCOA4 significantly attenuated the regulatory effect of 2-pyridylhydrazone dithiocarbamate s-acetic acid (PdtaA) on related proteins which highlighted that the strength of ferritinophagic flux (NCOA4/ferritin) was a driving force in determination of the status of EMT and ferroptosis. PdtaA treatment resulted in ferritinophagy that triggered ROS production in liver cancer cells. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [33] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Formosanin C | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model | Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Response regulation | The saponin formosanin C (FC) is a novel natural ferroptosis inducer, which triggered a stronger ferroptosis in human hepatocellular carcinoma HepG2 cells containing a higher level of NCOA4 and a lower level of FTH1 compared to Hep3B cells. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [34] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Regulator | Polypyrimidine tract-binding protein 1 (PTBP1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
In Vivo Model |
A total of eight male BALB/c nude mice (4-5 weeks old; weight, 13-18 g) were obtained from Beijing Vital River Laboratories. The mice were randomly divided into the following two groups: The NC + SF and sh-PTBP1 + SF groups. Hep3B cells (2 x 106) were subcutaneously injected into the right flank of each mouse. The mice were injected intraperitoneally with SF (10 mg/kg) every 2 days for 2 weeks.
Click to Show/Hide
|
||||
Response regulation | PTBP1 mediates ferroptosis in liver cancer cells by regulating NCOA4 translation.In vivoexperiments reconfirmed the role of the PTBP1NCOA4axis in a xenograft transplantation model. It was observed that the mean tumor weight increased afterPTBP1knockout. | ||||
Monocarboxylate transporter 1 (SLC16A1)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Lactate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
AMPK signaling pathway | hsa04152 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | CAF cells | Normal | Carassius auratus | CVCL_R883 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
In Vivo Model |
Female mice aged around 6-7 weeks were used for this study, which were purchased through Laboratory Animal Center of Chongqing Medical University from Vital River Co. Ltd (Beijing, China).After one week, each mouse was injected subcutaneously with 100 uL of Huh-7 cell suspension (5 x 106 units) to establish the tumor model. The mice were grouped randomly, and then subjected to different treatments after subcutaneous tumors became visually detectable.
Click to Show/Hide
|
||||
Response regulation | Lactate regulates the ferroptosis of hepatocellular carcinoma cells. And blocking the lactate uptake via hydroxycarboxylic acid receptor 1 (HCAR1)/MCT1 (SLC16A1) inhibition promotes ferroptosis by activating the AMPK to downregulate SCD1, which may synergize with its acyl-coenzyme A synthetase 4 (ACSL4)-promoting effect to amplify the ferroptotic susceptibility. | ||||
Heme oxygenase 1 (HMOX1)
In total 2 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [39] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | Eupalinolide B | Investigative | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell migration | |||||
Cell proliferation | |||||
In Vitro Model | SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | |
HCCLM3 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_6832 | ||
In Vivo Model |
Four-week-old nude mice (BALB/c) were used, and they were housed for 1 week in a specific pathogen-free (SPF) environment before experimentation. Each mouse received subcutaneous injections of 1 x 106 SMMC-7721 or HCCLM3 cells in 200 uL phosphate-buffered saline (PBS) into both flanks. One group was intraperitoneally injected every two days with EB at 25 or 50 mg/mouse body weight, for a total of 3 weeks. As a control, DMSO was used in another group. Tumor size was measured to calculate tumor volume, and mouse body weights were monitored every two days.
Click to Show/Hide
|
||||
Response regulation | Eupalinolide B (EB) exerts anti-proliferative activity in hepatic carcinoma by blocking cell cycle arrest at S phase and inducing ferroptosis mediated by endoplasmic reticulum (ER) stress, as well as HO-1 activation. And EB has the ability to inhibit cell proliferation and migration in hepatic carcinoma. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [40] | ||||
Target for Ferroptosis | Driver/Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Haloperidol | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Response regulation | The study demonstrated that haloperidol strengthened sorafenib-induced ferroptosis in Hepatocellular carcinoma cell lines for the first time. During ferroptosis, haloperidol substantially increased the cellular levels of Fe2+, GSH and lipid peroxidation. Additionally, haloperidol induced the expression of HO-1. | ||||
Glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [41] | ||||
Target for Ferroptosis | Marker/Driver | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
In Vivo Model |
A total of 32 male BALB/c nude mice (age, 6-8 weeks; weight, 18-20 g) were obtained from HFK Bioscience Co. Ltd. and housed in a specific pathogen-free facility. The mice were randomly divided into four groups (n = 8/group) and PLC cells were subcutaneously injected into the nude mice. When the xenografted tumors had grown to 80-100 mm3, half of the mice in each group were administered with 100 mg/kg DHA for 5 days/week by gavage.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin triggers ferroptosis in primary liver cancer cells by promoting and unfolded protein responseinduced upregulation of CHAC1 expression. DHA also promoted the transcription of CHAC1. | ||||
Ferroptosis suppressor protein 1 (AIFM2)
In total 2 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [42] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Vigilin (HDLBP) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
SNU-387 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0250 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
Six-week-old female BALB/c nude mice were purchased from Byrness Weil Biotechnology Ltd. (Chengdu, China) and housed in a specific pathogen-free environment with a 12-h light/dark cycle and controlled temperature and humidity, and food and water were provided ad libitum. Three million designated treated PLC5 cells were collected and injected subcutaneously into mice. At least 4 mice were used in each group in each experiment. Once the tumours reached a mean volume of 200 mm3, the mice were treated intraperitoneally with sorafenib every 3 days. The mice were then euthanized at the indicated time after injection. Each tumour was dissected, fixed with 4% formaldehyde, and embedded in paraffin. The tumour growth was monitored weekly by calliper measurements.
Click to Show/Hide
|
||||
Response regulation | LncFAL reduced ferroptosis vulnerability by directly binding to ferroptosis suppressor protein 1 (FSP1) and competitively abolishing Trim69-dependent FSP1 polyubiquitination degradation. Collectively, our results provide a clinically promising demonstration that HDLBP stabilizes lncFAL, which mediates a FSP1-dependent anti-ferroptosis mechanism in Hepatocellular carcinoma. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [42] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | lncFAL (IncRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
SNU-387 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0250 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
Six-week-old female BALB/c nude mice were purchased from Byrness Weil Biotechnology Ltd. (Chengdu, China) and housed in a specific pathogen-free environment with a 12-h light/dark cycle and controlled temperature and humidity, and food and water were provided ad libitum. Three million designated treated PLC5 cells were collected and injected subcutaneously into mice. At least 4 mice were used in each group in each experiment. Once the tumours reached a mean volume of 200 mm3, the mice were treated intraperitoneally with sorafenib every 3 days. The mice were then euthanized at the indicated time after injection. Each tumour was dissected, fixed with 4% formaldehyde, and embedded in paraffin. The tumour growth was monitored weekly by calliper measurements.
Click to Show/Hide
|
||||
Response regulation | LncFAL reduced ferroptosis vulnerability by directly binding to ferroptosis suppressor protein 1 (FSP1) and competitively abolishing Trim69-dependent FSP1 polyubiquitination degradation. Collectively, our results provide a clinically promising demonstration that HDLBP stabilizes lncFAL, which mediates a FSP1-dependent anti-ferroptosis mechanism in Hepatocellular carcinoma. | ||||
Ferritin light chain (FTL)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [18] | |||
Target for Ferroptosis | Suppressor | |||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | |||
Responsed Drug | Atractylodin | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Apoptosis | hsa04210 | |||
Cell Process | Cell ferroptosis | |||
Cell apoptosis | ||||
Cell proliferation | ||||
Cell migration | ||||
Cell invasion | ||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 |
Hccm (Human hepatocellular carcinoma cells) | ||||
Response regulation | Atractylodin can inhibit the proliferation, migration, and invasion of Huh7 and Hccm liver cancer cells, and induce cell apoptosis and cell cycle arrest. In addition, atractylodin may induce ferroptosis in hepatocellular carcinoma cells by inhibiting the expression of GPX4 and FTL proteins, and up-regulating the expression of ACSL4 and TFR1 proteins. | |||
Ferritin heavy chain (FTH1)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [33] | |||
Target for Ferroptosis | Marker/Suppressor | |||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | |||
Responsed Drug | Formosanin C | Investigative | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Autophagy | hsa04140 | |||
Cell Process | Cell ferroptosis | |||
Cell autophagy | ||||
In Vitro Model | Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Response regulation | The saponin formosanin C (FC) is a novel natural ferroptosis inducer, which triggered a stronger ferroptosis in human hepatocellular carcinoma HepG2 cells containing a higher level of NCOA4 and a lower level of FTH1 compared to Hep3B cells. | |||
Cystine/glutamate transporter (SLC7A11)
In total 16 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [43] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Necrostatin-1 | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
Response regulation | Necrostatin-1 potentiated sulfasalazine-induced expression of SLC7A11, a catalytic subunit of system xc- in these cells. And necrostatin-1 Prevents Ferroptosis in a RIPK1- and IDO-Independent Manner in Hepatocellular Carcinoma. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [12] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Drug | Sorafenib | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
SNU-182 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0090 | ||
SNU-449 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0454 | ||
In Vivo Model |
A total of 20 male Balb/c nude mice aged 6-8 weeks were purchased from Hunan SJA Laboratory Animal Co., Ltd. (Changsha, China). Five million Huh7 cells were inoculated into the right flanks of the mice. When the tumor size reached 80-100 mm3, the mice were randomly divided into four groups and administered artesunate (30 mg/kg mouse weight) alone, sorafenib (20 mg/kg mouse weight) alone, a combination of artesunate and sorafenib, or the same volume of PBS by gavage every other day.
Click to Show/Hide
|
||||
Response regulation | Sorafenib at low dose mainly caused oxidative stress through mitochondrial impairments and SLC7A11-invovled glutathione depletion. Artesunate-induced lysosome activation synergized with sorafenib-mediated pro-oxidative effects by promoting sequential reactions including lysosomal cathepsin B/L activation, ferritin degradation, lipid peroxidation, and consequent ferroptosis. Taken together, artesunate could be repurposed to sensitize sorafenib in hepatocellular carcinoma treatment. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [44] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | RNA-binding protein PNO1 (PNO1) | Suppressor | |||
Pathway Response | Autophagy | hsa04140 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
In Vivo Model |
Xenograft mouse model experiments were used male BALB/c nude mice (4 weeks old) purchased from SPF Biotechnology (Beijing, China). Each mouse was injected 5 x 106 tumor cells at the volume of 100 uL into the subcutaneous tissue. The tumor volume and weight of the mice was observed every 2 days. Mice were monitored daily and the tumor volume calculated according to the equation volume = length x width2 x 1/2.
Click to Show/Hide
|
||||
Response regulation | PNO1 inhibits autophagy-mediated ferroptosis via GSH metabolic reprogramming as demonstrated above. We also demonstrated that PNO1 inhibition repressed SLC7A11 through p53 to promote ferroptosis. These observations suggested that sh-PNO1 could be a new target in hepatocellular carcinoma therapy. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [45] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | ATP-binding cassette sub-family C member 5 (ABCC5) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
In Vivo Model |
All animal experiments were carried out with the approval of the Southern Medical University Animal Care and Use Committee in accordance with the guidelines for the ethical treatment of animals. Nude nu/nu mice were maintained in a barrier facility in racks filtered with a high-efficiency particulate air filter. The animals were fed an autoclaved laboratory rodent diet. The mice in this study were purchased from the Experimental Animal Centre of Southern Medical University, which is certified by the Guangdong Provincial Bureau of Science.
Click to Show/Hide
|
||||
Response regulation | ABCC5 increased intracellular glutathione (GSH) and attenuated lipid peroxidation accumulation by stabilizing SLC7A11 protein, which inhibited ferroptosis. Additionally, the inhibition of ABCC5 enhanced the anti-cancer activity of sorafenib in hepatocellular carcinoma. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [46] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Transforming growth factor beta-1 proprotein (TGFB1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
SNU-387 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0250 | ||
SNU-449 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0454 | ||
SNU475 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0497 | ||
SK-HEP-1 cells | Liver and intrahepatic bile duct epithelial neoplasm | Homo sapiens | CVCL_0525 | ||
HuH-6 cells | Hepatoblastoma | Homo sapiens | CVCL_4381 | ||
Response regulation | TGF-B1 represses xCT (SLC7A11) expression via Smad3 activation and enhances lipid peroxidation in hepatocellular carcinoma cells with an early TGF-B1 signature, which would benefit from the targeting of GPX4. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [47] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Cyclic AMP-dependent transcription factor ATF-4 (ATF4) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Hippo signaling pathway | hsa04390 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
SNU-398 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0077 | ||
HT-1080 cells | Fibrosarcoma | Homo sapiens | CVCL_0317 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
HLE cells | Hepatocellular carcinoma | Homo sapiens | CVCL_1281 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
SNU398-parental cells, SNU398-shLuc, or SNU398-shYAP/TAZ cells (106 in 100 ul PBS) were implanted into the left flanks of immunodeficient NOD/SCID; common receptor-/-(NSG) mice. When tumors were palpable, Sorafenib (LC Laboratories, S-8502) was applied at 20 mg/kg daily via gavage, SSA (Sulfasalazine, Sigma, S0883) was given at 120 mg/kg daily by intraperitoneal injection, 20 mM BSO (Lbuthionine-sulfoximine, Sigma, B2515) was given in the drinking water for 3 weeks.
Click to Show/Hide
|
||||
Response regulation | In a TEAD-dependent manner, YAP/TAZ induce the expression of SLC7A11, a key transporter maintaining intracellular glutathione homeostasis, thus enabling hepatocellular carcinoma cells to overcome Sorafenib-induced ferroptosis. At the same time, YAP/TAZ sustain the protein stability, nuclear localization, and transcriptional activity of ATF4 which in turn cooperates to induce SLC7A11 expression. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [47] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Tafazzin (TAFAZZIN) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Hippo signaling pathway | hsa04390 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
SNU-398 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0077 | ||
HT-1080 cells | Fibrosarcoma | Homo sapiens | CVCL_0317 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
HLE cells | Hepatocellular carcinoma | Homo sapiens | CVCL_1281 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
SNU398-parental cells, SNU398-shLuc, or SNU398-shYAP/TAZ cells (106 in 100 ul PBS) were implanted into the left flanks of immunodeficient NOD/SCID; common receptor-/-(NSG) mice. When tumors were palpable, Sorafenib (LC Laboratories, S-8502) was applied at 20 mg/kg daily via gavage, SSA (Sulfasalazine, Sigma, S0883) was given at 120 mg/kg daily by intraperitoneal injection, 20 mM BSO (Lbuthionine-sulfoximine, Sigma, B2515) was given in the drinking water for 3 weeks.
Click to Show/Hide
|
||||
Response regulation | In a TEAD-dependent manner, YAP/TAZ induce the expression of SLC7A11, a key transporter maintaining intracellular glutathione homeostasis, thus enabling hepatocellular carcinoma cells to overcome Sorafenib-induced ferroptosis. At the same time, YAP/TAZ sustain the protein stability, nuclear localization, and transcriptional activity of ATF4 which in turn cooperates to induce SLC7A11 expression. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [48] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | DAZ-associated protein 1 (DAZAP1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
Male BALB/c nude mice (4-5 weeks, 14-18 g) were purchased from Vital River Laboratories (Beijing, China). We randomly (random number grouping method) divided the mice into five groups: the blank group, the DMSO group, the SF group, the SF + sh-NC group and the SF + sh-DAZAP1 group. For SF-intervention mice, we dissolved 10 mg/kg of SF into 0.2 ml DMSO and injected the mixture intraperitoneally every other day for two weeks. 1 x 107 cells (with or without lentivirus) suspended in 500 ul of ice-cold PBS were subcutaneously injected into the left flank of mice.
Click to Show/Hide
|
||||
Response regulation | DAZAP1 knockdown by small interfering RNA markedly inhibited hepatocellular carcinoma (HCC) cell proliferation, migration and invasion. At the mechanistic level, DAZAP1 was identified as a potent inhibitor of ferroptosis and an efficient binding partner of SLC7A11 mRNA. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [49] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | N6-adenosine-methyltransferase non-catalytic subunit (METTL14) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Glutamate metabolism | hsa00250 | ||||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
SMMC-7721 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0534 | ||
HCCLM3 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_6832 | ||
MHCC97-H cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_4972 | ||
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
In Vivo Model |
The BALB/C nude mice were obtained from Shanghai SLAC Laboratory Animal Co., Ltd. 5 x 105 stable SLC7A11-knockdown HCCLM3 cells or SLC7A11-vector cells were injected subcutaneously into BALB/C nude mice. After 5 weeks, mice were killed, and tumour photograph was detected with photography. In the other animal work, 5 x 105 stable METTL14-vector HCCLM3 cells, SLC7A11-Overexpression cells or SLC7A11-R298P cells were injected subcutaneously into BALB/C nude mice.
Click to Show/Hide
|
||||
Response regulation | METTL14 induced m6A modification at 5'UTR of SLC7A11 mRNA, which in turn underwent degradation relied on the YTHDF2-dependent pathway. Importantly, ectopic expression of SLC7A11 strongly blocked METTL14-induced tumour-suppressive effect in hypoxic hepatocellular carcinoma. | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [44] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | RNA-binding protein PNO1 (PNO1) | Suppressor | |||
Pathway Response | Autophagy | hsa04140 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
In Vivo Model |
Xenograft mouse model experiments were used male BALB/c nude mice (4 weeks old) purchased from SPF Biotechnology (Beijing, China). Each mouse was injected 5 x 106 tumor cells at the volume of 100 uL into the subcutaneous tissue. The tumor volume and weight of the mice was observed every 2 days. Mice were monitored daily and the tumor volume calculated according to the equation volume = length x width2 x 1/2.
Click to Show/Hide
|
||||
Response regulation | PNO1 inhibits autophagy-mediated ferroptosis via GSH metabolic reprogramming as demonstrated above. We also demonstrated that PNO1 inhibition repressed SLC7A11 through p53 to promote ferroptosis. These observations suggested that sh-PNO1 could be a new target in Hepatocellular carcinoma therapy. | ||||
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target | [47] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Transcriptional coactivator YAP1 (YAP1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Hippo signaling pathway | hsa04390 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
SNU-398 cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_0077 | ||
HT-1080 cells | Fibrosarcoma | Homo sapiens | CVCL_0317 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
HLE cells | Hepatocellular carcinoma | Homo sapiens | CVCL_1281 | ||
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | ||
In Vivo Model |
SNU398-parental cells, SNU398-shLuc, or SNU398-shYAP/TAZ cells (106 in 100 ul PBS) were implanted into the left flanks of immunodeficient NOD/SCID; common receptor-/-(NSG) mice. When tumors were palpable, Sorafenib (LC Laboratories, S-8502) was applied at 20 mg/kg daily via gavage, SSA (Sulfasalazine, Sigma, S0883) was given at 120 mg/kg daily by intraperitoneal injection, 20 mM BSO (Lbuthionine-sulfoximine, Sigma, B2515) was given in the drinking water for 3 weeks.
Click to Show/Hide
|
||||
Response regulation | In a TEAD-dependent manner, YAP/TAZ induce the expression of SLC7A11, a key transporter maintaining intracellular glutathione homeostasis, thus enabling hepatocellular carcinoma cells to overcome Sorafenib-induced ferroptosis. At the same time, YAP/TAZ sustain the protein stability, nuclear localization, and transcriptional activity of ATF4 which in turn cooperates to induce SLC7A11 expression. | ||||
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target | [50] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Transcriptional coactivator YAP1 (YAP1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Hippo signaling pathway | hsa04390 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell metastasis | |||||
In Vitro Model | Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
MHCC97-H cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_4972 | ||
HCC-LY10 (Human hepatoma cells) | |||||
In Vivo Model |
To generate murine subcutaneous tumors, 2 x 106 HCC cells were injected subcutaneously to the right of the dorsal midline in nude mice. Once the tumors reached approximately 100 mm3 at day 15, mice were randomly allocated into groups and treated with erastin or sorafenib for 2 weeks.
Click to Show/Hide
|
||||
Response regulation | Overexpression of AKR1C3 protected against ferroptosis in hepatocellular carcinoma (HCC) cells. Mechanistically, AKR1C3 regulated ferroptosis through YAP/SLC7A11 signaling in HCC. | ||||
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target | [50] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Aldo-keto reductase family 1 member C3 (AKR1C3) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Hippo signaling pathway | hsa04390 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell metastasis | |||||
In Vitro Model | Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | ||
MHCC97-H cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_4972 | ||
HCC-LY10 (Human hepatoma cells) | |||||
In Vivo Model |
To generate murine subcutaneous tumors, 2 x 106 HCC cells were injected subcutaneously to the right of the dorsal midline in nude mice. Once the tumors reached approximately 100 mm3 at day 15, mice were randomly allocated into groups and treated with erastin or sorafenib for 2 weeks.
Click to Show/Hide
|
||||
Response regulation | Overexpression of AKR1C3 protected against ferroptosis in hepatocellular carcinoma (HCC) cells. Mechanistically, AKR1C3 regulated ferroptosis through YAP/SLC7A11 signaling in HCC. | ||||
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target | [51] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | hsa-miR-1261 (miRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell invasion | |||||
Cell metastasis | |||||
In Vitro Model | L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
MHCC97-H cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_4972 | ||
In Vivo Model |
Four-week-old female BALB/c nude mice were subcutaneously inoculated with 2 x 106 cells (5 mice per group). Intratumoral injection (40 uL of si-circ0097009 or negative control siRNA) was administered every 4 days. Mice were sacrificed, and tumor weights were measured after 4 weeks. To establish lung metastases, HepG2 cells were treated with 20 umol/L si-circ0097009. After 48 hours, cells (1 x 105) were intravenously injected into the tail veins of mice (6 mice per group).
Click to Show/Hide
|
||||
Response regulation | Circ0097009 acts as a competing endogenous RNA to regulate the expression of SLC7A11, a key regulator of cancer cell ferroptosis, by sponging miR-1261 in hepatocellular cancer. Circ0097009 may be used as a diagnostic biomarker for HCC and as a potential target for HCC therapy. | ||||
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target | [31] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | HEPFAL (IncRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | ||
Response regulation | LncRNA HEPFAL promotes the ubiquitination of SLC7A11, resulting in a decrease in GSH production, which in turn affects the activity of GPX4 and ultimately leads to the occurrence of ferroptosis. And LncRNA HEPFAL has the potential as a target for the diagnosis and treatment of hepatocellular carcinoma. | ||||
Experiment 16 Reporting the Ferroptosis-centered Disease Response by This Target | [51] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | ||||
Responsed Regulator | Circ0097009 (circRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell invasion | |||||
Cell metastasis | |||||
In Vitro Model | L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
MHCC97-H cells | Adult hepatocellular carcinoma | Homo sapiens | CVCL_4972 | ||
In Vivo Model |
Four-week-old female BALB/c nude mice were subcutaneously inoculated with 2 x 106 cells (5 mice per group). Intratumoral injection (40 uL of si-circ0097009 or negative control siRNA) was administered every 4 days. Mice were sacrificed, and tumor weights were measured after 4 weeks. To establish lung metastases, HepG2 cells were treated with 20 umol/L si-circ0097009. After 48 hours, cells (1 x 105) were intravenously injected into the tail veins of mice (6 mice per group).
Click to Show/Hide
|
||||
Response regulation | Circ0097009 acts as a competing endogenous RNA to regulate the expression of SLC7A11, a key regulator of cancer cell ferroptosis, by sponging miR-1261 in hepatocellular cancer. Circ0097009 may be used as a diagnostic biomarker for Hepatocellular carcinoma and as a potential target for HCC therapy. | ||||
4F2 cell-surface antigen heavy chain (SLC3A2)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [52] | |||
Target for Ferroptosis | Suppressor | |||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | |||
Responsed Regulator | hsa-miR-142-3p (miRNA) | Driver | ||
Pathway Response | Glutathione metabolism | hsa00480 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
Cell migration | ||||
Cell invasion | ||||
In Vitro Model | Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 |
THP-1 cells | Childhood acute monocytic leukemia | Homo sapiens | CVCL_0006 | |
Response regulation | MiR-142-3p promoted HBV-infected M1-type macrophage ferroptosis through SLC3A2, affecting the production of GSH, MDA, and Fe2+and accelerating the development of hepatocellular carcinoma (HCC). Inhibition of miR-142-3p or overexpression of SLC3A2 reversed ferroptosis and inhibited the proliferation, migration, and invasion of HCC cells. | |||
CDGSH iron-sulfur domain-containing protein 2 (CISD2)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [69] | |||
Target for Ferroptosis | Driver/Suppressor | |||
Responsed Disease | Hepatocellular carcinoma [ICD-11: 2C12.02] | |||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Autophagy | hsa04140 | |||
Cell Process | Cell ferroptosis | |||
Cell autophagy | ||||
In Vitro Model | L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 |
Hep 3B2.1-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0326 | |
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | |
Huh-7 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0336 | |
PLC/PRF/5 cells | Hepatocellular carcinoma | Homo sapiens | CVCL_0485 | |
Response regulation | Inhibition of CISD2 promoted sorafenib-induced ferroptosis in resistant cells, and this process promoted excessive iron ion accumulation through autophagy, leading to ferroptosis. The combination of CISD2 inhibition and sorafenib treatment is an effective therapeutic strategy for resistant hepatocellular carcinoma (HCC). | |||
References