General Information of the Disease (ID: DIS00147)
Name
Injury of intra-abdominal organs
ICD
ICD-11: NB91
Full List of Target(s) of This Ferroptosis-centered Disease
Nuclear factor erythroid 2-related factor 2 (NFE2L2)
In total 12 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Ulinastatin Phase 3
Responsed Regulator NAD-dependent protein deacetylase sirtuin-1 (SIRT1) Suppressor
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
Male C57BL/6 mice were from the Experimental Animal Center of Xian Jiaotong University. The animal experiment procedures were performed in accordance with the Guide of Laboratory Animal Care and Use from the United States National Institution of Health and were approved by the Laboratory Animal Care Committee (LACC) of Xian Jiaotong University, China (No. XJTULAC2017-207). Mice were initially housed for 7 days to adjust to the environment. The experimental design included five groups (n = 10 per group): the control group included the saline control (0.9% saline) group, and the test groups included APAP, APAP + UTI (5 x 104 units/kg and 1 x 105 units/kg), APAP + Fer-1 (10 mg/kg), and APAP + Res (50 mg/kg) treatments administered by tail vein or intraperitoneal injection.

    Click to Show/Hide
Response regulation Ulinastatin plays a role in mitigation of Acetaminophen (APAP)-induced acute liver injury by inhibiting ferroptosis-induced lipid peroxide accumulation, and the effect of UT1 was mediated by the NRF2/HO-1 pathway and SIRT1 expression.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Drug Baicalin Terminated
Responsed Regulator Kelch-like ECH-associated protein 1 (KEAP1) Driver
Pathway Response Pathways in cancer hsa05200
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vivo Model
C57BL/6 mice at 6-8 weeks were intraperitoneally injected with D-GalN/LPS (1772-03-8/L2880, Sigma-Aldrich, USA) at a dose of 700 mg/kg and 10 ug/kg, respectively. The constructed D-GaIN/LPS-induced ALI model mice were named the model group, and the normal mice injected with phosphate-buffered saline (PBS) were named the blank group. After 1 h of LPS/D-GalN treatment, Exo and Ba-Exo (150 ug/mice) were injected into the tail vein of the mice in the Exo and Ba-Exo groups, respectively. Mice were sacrificed via anesthesia overdose 12 h after the intervention. Half of the liver tissue was fixed in paraformaldehyde, while the other half was frozen at 80 . Peripheral blood serum was stored at -80 .

    Click to Show/Hide
Response regulation Baicalin-pretreated MSCs (Ba-Exo) exerts a protective effect on liver function and activates the Keap1-NRF2 pathway via P62 (SQSTM1), thereby inhibiting ROS production and lipid peroxide-induced ferroptosis. Therefore, baicalin pretreatment is an effective and promising approach in optimizing the therapeutic efficacy of Exo in acute liver injury (ALI).
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Drug Baicalin Terminated
Responsed Regulator Sequestosome-1 (SQSTM1) Driver
Pathway Response Pathways in cancer hsa05200
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vivo Model
C57BL/6 mice at 6-8 weeks were intraperitoneally injected with D-GalN/LPS (1772-03-8/L2880, Sigma-Aldrich, USA) at a dose of 700 mg/kg and 10 ug/kg, respectively. The constructed D-GaIN/LPS-induced ALI model mice were named the model group, and the normal mice injected with phosphate-buffered saline (PBS) were named the blank group. After 1 h of LPS/D-GalN treatment, Exo and Ba-Exo (150 ug/mice) were injected into the tail vein of the mice in the Exo and Ba-Exo groups, respectively. Mice were sacrificed via anesthesia overdose 12 h after the intervention. Half of the liver tissue was fixed in paraformaldehyde, while the other half was frozen at 80 . Peripheral blood serum was stored at -80 .

    Click to Show/Hide
Response regulation Baicalin-pretreated MSCs (Ba-Exo) exerts a protective effect on liver function and activates the Keap1-NRF2 pathway via P62 (SQSTM1), thereby inhibiting ROS production and lipid peroxide-induced ferroptosis. Therefore, baicalin pretreatment is an effective and promising approach in optimizing the therapeutic efficacy of Exo in acute liver injury (ALI).
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [3]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Acetaminophen-induced liver injury [ICD-11: DB95.Z]
Responsed Drug Clausenamide Investigative
Responsed Regulator Kelch-like ECH-associated protein 1 (KEAP1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Pathways in cancer hsa05200
Cell Process Cell ferroptosis
In Vitro Model HepaRG cells Hepatocellular carcinoma Homo sapiens CVCL_9720
SMMC-7721 cells Endocervical adenocarcinoma Homo sapiens CVCL_0534
Hep-G2 cells Hepatoblastoma Homo sapiens CVCL_0027
BEL-7402 cells Endocervical adenocarcinoma Homo sapiens CVCL_5492
In Vivo Model
Male C57BL/6 mice aged 8-10 weeks were purchased from Guangdong Experimental Animal Center (Guangzhou, China). The animals were maintained on a 12 h light-dark cycle in a regulated temperature and humidity environment for 1 week before drug administration. (+)-CLA (50 mg/kg/day, i.g.) or fer-1 (2.5 umol/kg/day, i.p.)were administered for 7 consecutive days. To induce liver injury, mice were injected with erastin (100 mg/kg/day, i.p., twice a day) on both the 6th and 7th day, or a single dose of APAP (600 mg/kg/day, i.p.) on the 7th day after overnight food deprivation. The serum and livers were obtained for analysis.

    Click to Show/Hide
Response regulation (+)-clausenamide ((+)-CLA) specifically reacted with the Cys-151 residue of Keap1, which blocked Nrf2 ubiquitylation and resulted in an increased Nrf2 stability. Thus, (+)-CLA protects against acetaminophen-induced hepatotoxicity via inhibiting ferroptosis and activating the Keap1/Nrf2 pathway in a Cys-151-dependent manner.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [4]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Xiaojianzhong Investigative
Responsed Regulator Kelch-like ECH-associated protein 1 (KEAP1) Driver
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model hGCs (Gastric cells)
In Vivo Model
C57BL/6 mice (Male, 6-8 weeks old, 20 g± 2 g) were purchased from Chengdu Yaokang Biotechnology Co., Ltd. (Chengdu, China). All animals were housed in the animal room of Shaanxi University ofTraditional Chinese Medicine, at a temperature of 22 ± 2 and a humidity of 40% ± 5%, alternating between light and dark. In the study, the mice were randomly divided into six groups (n = 10 in each group): the blank group, model group,XJZ high dose group, XJZ medium dose group, XJZ low dose group, and positive control (omeprazole) group. The mice in the model group were given Aspirin (300 mg/kg) via gavage for 14 days; the mice in the XJZ high dose group, XJZ medium dose group, and XJZ low dose group were given aspirin (300 mg/kg) by gavage in the morning and three different concentrations (12 g/kg, 6 g/kg, or 3 g/kg) of XJZ decoction by gavage in the afternoon; the mice in the positive control group were given aspirin (300 mg/kg) by gavage in the morning andomeprazole(20 mg/kg) by gavage in the afternoon. After the model was successfully constructed, the mice were anesthetized with isoflurane and gastric tissues were extracted for analysis.

    Click to Show/Hide
Response regulation Xiaojianzhong (XJZ) significantly counteracted aspirin-induced gastric mucosal injury and inhibited oxidative stress and ferroptosis in mice. Upon examining SQSTM1/p62(p62)/ Kelch-like ECH-associated protein 1 (Keap1)/Nuclear Factor erythroid 2-Related Factor 2 (Nrf2), a well-known signaling pathway involved in the regulation of oxidative stress and ferroptosis, we found that its activation was significantly inhibited by aspirin treatment and that this signaling pathway was activated after XJZ intervention.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Ulinastatin Phase 3
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
Male C57BL/6 mice were from the Experimental Animal Center of Xian Jiaotong University. The animal experiment procedures were performed in accordance with the Guide of Laboratory Animal Care and Use from the United States National Institution of Health and were approved by the Laboratory Animal Care Committee (LACC) of Xian Jiaotong University, China (No. XJTULAC2017-207). Mice were initially housed for 7 days to adjust to the environment. The experimental design included five groups (n = 10 per group): the control group included the saline control (0.9% saline) group, and the test groups included APAP, APAP + UTI (5 x 104 units/kg and 1 x 105 units/kg), APAP + Fer-1 (10 mg/kg), and APAP + Res (50 mg/kg) treatments administered by tail vein or intraperitoneal injection.

    Click to Show/Hide
Response regulation Ulinastatin plays a role in mitigation of APAP-induced acute liver injury by inhibiting ferroptosis-induced lipid peroxide accumulation, and the effect of UT1 was mediated by the NRF2/HO-1 pathway and SIRT1 expression.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [16]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Acetaminophen-induced liver injury [ICD-11: DB95.Z]
Responsed Drug Abietic acid Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model AML12 cells Normal Mus musculus CVCL_0140
In Vivo Model
APAP-induced liver injury model was induced byintraperitoneal injection 300 mg/kg APAP. The mice of APAP + abietic acid (10, 20, 40 mg/kg) were given abietic acid by intraperitoneal injection 1 h before APAP treatment. The doses of abietic acid used in this study were based on previous studies. Twelve hours later, the mice were sacrificed after anesthesia with 1%pentobarbital (50 mg/kg) injected intraperitoneally and the samples were collected.

    Click to Show/Hide
Response regulation APAP could increase malondialdehyde (MDA) and Fe2+ levels, and decrease ATP and glutathione (GSH) levels, as well as glutathione peroxidase 4 (GPX4) and xCT expression. However, these changes induced by APAP were prevented by abietic acid, indicating abietic acid could inhibit APAP-induced ferroptosis. Furthermore, abietic acid inhibited APAP-induced liver injury, NF-B activation and increased the expression of Nrf2 and HO-1.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Kaempferol Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
Male BALB/c mice (8-week-old, 20-22 g) were purchased from Guangdong Medical Laboratory Animal Center (Guangzhou, China). The experimental animals were fed adaptively for one week in the Experimental Animal Center of Guangdong Pharmaceutical University (Guangzhou, China). Feeding conditions were set at 26 , humidity 65% and a lightdark cycle for 12 hours. All animal experiments were performed following the Guide for the Care and Use of Laboratory Animals, and the procedures were approved by the Research Ethical Committee of Guangdong Pharmaceutical University (gdpulacspf2020007).

    Click to Show/Hide
Response regulation Kaempferol (KA) activated the Nrf2 pathway and upregulated Gpx4 in mouse livers and L02 cells to inhibit ferroptosis induced by APAP. Finally, molecular docking indicated the potential interaction of KA with Keap1. Taken together, KA ameliorated oxidative stress and ferroptosis-mediated acetaminophen-induced liver injury by activating Nrf2 signaling.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [17]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Acetaminophen-induced liver injury [ICD-11: DB95.Z]
Responsed Drug Astaxanthin Investigative
Pathway Response NF-kappa B signaling pathway hsa04064
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell autophagy
Cell apoptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
Mice were randomly divided into four groups as follows (n = 5): (1) control, (2) APAP (MCE, Monmouth Junction, NJ, USA), (3) olive oil + APAP (oil + APAP), and (4) ASX (Energy Chemical, Shanghai, China) dissolved in olive oil + APAP (ASX + APAP). Astaxanthin was dissolved in olive oil to obtain a mixture of 20 mg/mL. Mice in groups 3 and 4 were given a dose of olive oil and a mixture of 5 mL/kgBW by gavage every day for 2 weeks. On day 15, mice in groups 2, 3, and 4 were given a peritoneal injection of 500 mg/kg APAP to induce liver injury. The mice were fasted for 12 h before the administration of APAP. Ten hours after APAP administration, blood and liver tissue were collected for further examination and analyses. Blood was centrifuged to obtain supernatants,which were stored at -80. Liver tissues were immediately removed from each animal, and homogenates were processed with formaldehyde and glutaraldehyde for protein and histological analysis.

    Click to Show/Hide
Response regulation Astaxanthin reduced inflammation through the NF-B pathway, inhibited oxidative stress and ferroptosis, and increased autophagy through the Nrf2/HO-1 pathway, ameliorating acetaminophen-induced liver injury in vivo and in vitro.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Target for Ferroptosis Marker/Suppressor
Responsed Disease CCl4-induced iver injury [ICD-11: NB91]
Responsed Drug Bicyclol Investigative
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
The mice were treated with intraperitoneal administration (i.p.) of oil (control group) or a mixture of CCl4 (50%) and oil (50%) at a dosage of 2 ml/kg body weight. In the bicyclol-treated group, mice accepted administration of 200 mg/kg (using 0.5% carboxymethyl cellulose as solvent) by gavage three times a day 1 h before CCl4 exposure, while other groups accepted vehicles of the equal volume. Fer-1 was prepared in DMSO (5 mg/kg), andi.p. injected into mice once 1 h before CCl4 exposure. The dosage of bicyclol was consistent with our previous work. The mice were then sacrificed to collect liver and serum samples after 24 or 48 h.

    Click to Show/Hide
Response regulation Bicyclol exerted its hepatoprotection by preventing the aforesaid ferroptotic process. Furthermore, bicyclol alleviated erastin-induced cellular inviability, destruction, and lipid peroxidation in vitro. Knockdown of GPX4 diminished these protective activities against perturbations associated with ferroptosis in L-O2 hepatocytes. Additionally, Nrf2 silencing drastically reduced GPX4 levels, and further impeded the medicinal effects of bicyclol. In summary, positively regulating Nrf2-GPX4 axis by bicyclol can prevent ferroptosis in CCl4-induced acute liver injury in mice.
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target [13]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Liver Oxidative Damage [ICD-11: NB91]
Responsed Drug Epigallocatechin Gallate Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
In Vivo Model
All mice were randomly divided into a 2 x 2 factorial arrangement, fed diets containing 40 mg/kg or 5000 mg/kg FeSO4 (the basis of the diet was AIN-93), and gavaged with PBS or 50 mg EGCG/kg body weight per day, respectively. The experiment lasted for 6 weeks, including a 1-week adaptation and a 3-week EGCG gavage; then, all mice were euthanized.

    Click to Show/Hide
Response regulation Epigallocatechin-3-Gallate (EGCG) supplementation alleviated the liver oxidative damage caused by iron overload by inhibiting ferroptosis. EGCG addition increased NRF2 and GPX4 expression and elevated antioxidant capacity in iron overload mice. EGCG administration attenuates iron metabolism disorders by upregulating FTH/FTL expression. Through these two mechanisms, EGCG can effectively inhibit iron overload-induced ferroptosis.
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target [18]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Regulator RelA-associated inhibitor (PPP1R13L) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model MLE-2 (Mouse lung epithelial cells)
mLT (Mouse lung tissue)
In Vivo Model
8-week-old Nrf2-knockout (Nrf2-/-) and wild-type (WT) littermate male mice on a C57BL/6J background (provided by the RIKEN Bio-Resource Center through the National Bio-Resource Project of MEXT, Japan) were used for in vivo experiments. Intestinal ischemia was induced by clamping of the superior mesenteric artery after the intraperitoneal injection of 50 mg/kg of sodium pentobarbital. After 90 min, the intestine was reperfused for the times indicated. Sham control mice underwent the same procedure without vascular occlusion.

    Click to Show/Hide
Response regulation iASPP (PPP1R13L) mediated its protective effects against acute lung injury through the Nrf2/HIF-1/TF signaling pathway. Ferroptosis contributes to intestinal ischemia/reperfusion-induced acute lung injury (ALI), and iASPP treatment inhibits ferroptosis in part via Nrf2.
Heme oxygenase 1 (HMOX1)
In total 4 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Acetaminophen-induced liver injury [ICD-11: DB95.Z]
Responsed Drug Ulinastatin Phase 3
Responsed Regulator NAD-dependent protein deacetylase sirtuin-1 (SIRT1) 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
In Vivo Model
Male C57BL/6 mice were from the Experimental Animal Center of Xian Jiaotong University. The animal experiment procedures were performed in accordance with the Guide of Laboratory Animal Care and Use from the United States National Institution of Health and were approved by the Laboratory Animal Care Committee (LACC) of Xian Jiaotong University, China (No. XJTULAC2017-207). Mice were initially housed for 7 days to adjust to the environment. The experimental design included five groups (n = 10 per group): the control group included the saline control (0.9% saline) group, and the test groups included APAP, APAP + UTI (5 x 104 units/kg and 1 x 105 units/kg), APAP + Fer-1 (10 mg/kg), and APAP + Res (50 mg/kg) treatments administered by tail vein or intraperitoneal injection.

    Click to Show/Hide
Response regulation Ulinastatin (UT1) plays a role in mitigation of Acetaminophen (APAP)-induced acute liver injury by inhibiting ferroptosis-induced lipid peroxide accumulation, and the effect of UT1 was mediated by the NRF2/HO-1 pathway and SIRT1 expression.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Ulinastatin Phase 3
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
Male C57BL/6 mice were from the Experimental Animal Center of Xian Jiaotong University. The animal experiment procedures were performed in accordance with the Guide of Laboratory Animal Care and Use from the United States National Institution of Health and were approved by the Laboratory Animal Care Committee (LACC) of Xian Jiaotong University, China (No. XJTULAC2017-207). Mice were initially housed for 7 days to adjust to the environment. The experimental design included five groups (n = 10 per group): the control group included the saline control (0.9% saline) group, and the test groups included APAP, APAP + UTI (5 x 104 units/kg and 1 x 105 units/kg), APAP + Fer-1 (10 mg/kg), and APAP + Res (50 mg/kg) treatments administered by tail vein or intraperitoneal injection.

    Click to Show/Hide
Response regulation Ulinastatin plays a role in mitigation of APAP-induced acute liver injury by inhibiting ferroptosis-induced lipid peroxide accumulation, and the effect of UT1 was mediated by the NRF2/HO-1 pathway and SIRT1 expression.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [16]
Target for Ferroptosis Suppressor
Responsed Disease Acetaminophen-induced liver injury [ICD-11: DB95.Z]
Responsed Drug Abietic acid Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model AML12 cells Normal Mus musculus CVCL_0140
In Vivo Model
APAP-induced liver injury model was induced byintraperitoneal injection 300 mg/kg APAP. The mice of APAP + abietic acid (10, 20, 40 mg/kg) were given abietic acid by intraperitoneal injection 1 h before APAP treatment. The doses of abietic acid used in this study were based on previous studies. Twelve hours later, the mice were sacrificed after anesthesia with 1%pentobarbital (50 mg/kg) injected intraperitoneally and the samples were collected.

    Click to Show/Hide
Response regulation APAP could increase malondialdehyde (MDA) and Fe2+ levels, and decrease ATP and glutathione (GSH) levels, as well as glutathione peroxidase 4 (GPX4) and xCT expression. However, these changes induced by APAP were prevented by abietic acid, indicating abietic acid could inhibit APAP-induced ferroptosis. Furthermore, abietic acid inhibited APAP-induced liver injury, NF-B activation and increased the expression of Nrf2 and HO-1.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [17]
Target for Ferroptosis Suppressor
Responsed Disease Acetaminophen-induced liver injury [ICD-11: DB95.Z]
Responsed Drug Astaxanthin Investigative
Pathway Response NF-kappa B signaling pathway hsa04064
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell autophagy
Cell apoptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
Mice were randomly divided into four groups as follows (n = 5): (1) control, (2) APAP (MCE, Monmouth Junction, NJ, USA), (3) olive oil + APAP (oil + APAP), and (4) ASX (Energy Chemical, Shanghai, China) dissolved in olive oil + APAP (ASX + APAP). Astaxanthin was dissolved in olive oil to obtain a mixture of 20 mg/mL. Mice in groups 3 and 4 were given a dose of olive oil and a mixture of 5 mL/kgBW by gavage every day for 2 weeks. On day 15, mice in groups 2, 3, and 4 were given a peritoneal injection of 500 mg/kg APAP to induce liver injury. The mice were fasted for 12 h before the administration of APAP. Ten hours after APAP administration, blood and liver tissue were collected for further examination and analyses. Blood was centrifuged to obtain supernatants,which were stored at -80. Liver tissues were immediately removed from each animal, and homogenates were processed with formaldehyde and glutaraldehyde for protein and histological analysis.

    Click to Show/Hide
Response regulation Astaxanthin reduced inflammation through the NF-B pathway, inhibited oxidative stress and ferroptosis, and increased autophagy through the Nrf2/HO-1 pathway, ameliorating acetaminophen-induced liver injury in vivo and in vitro.
Unspecific Target
In total 8 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Erastin Investigative
Responsed Regulator Sestrin-2 (SESN2) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model Hep-G2 cells Hepatoblastoma Homo sapiens CVCL_0027
AML12 cells Normal Mus musculus CVCL_0140
mEFs (Mouse embryonic fibroblasts)
In Vivo Model
Six-weeks-old male ICR mice were obtained from Orient Bio (Sungnam, Korea) and acclimatized for 1 week. For Sesn2 overexpression, ICR mice were injected with the recombinant adenovirus particles (1 x 109 pfu) suspended in phosphate-buffered saline with tail vein. After 48 h, phenylhydrazine (PHZ, 60 mg/kg, i.p.) was administered to induce iron accumulation and liver injury.

    Click to Show/Hide
Response regulation Treatment with erastin upregulated Sesn2 mRNA levels and luciferase reporter gene activity, and erastin-mediated Sesn2 induction was transcriptionally regulated by NF-E2-related factor 2 (Nrf2). Collectively, ferroptosis-mediated Sesn2 induction is dependent on Nrf2 and plays a protective role against iron overload and ferroptosis-induced liver injury.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [6]
Responsed Disease Intestinal injury [ICD-11: NB91]
Responsed Drug Microcystin-LR Investigative
Responsed Regulator Cyclic AMP-dependent transcription factor ATF-4 (ATF4) Driver
Pathway Response Glutathione metabolism hsa00480
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
In Vivo Model
In total, 90 common carp susceptible to MC-LR were obtained from Hulan Fishing Ground (Harbin, China) and acclimatized in 100 L glass aquaria (15 fish per tank) containing continuously aerated water at 23 under a 12 h light-dark cycle for 10 days prior to the experiments. Following acclimation, the fish in MC-LR treatment (M) group (45 fish in 3 tanks) were exposed to 10 ug/L of MC-LR for 15 days. The baseline mean body weights of the fish in the control (C) and M groups were 2.11 ± 0.03 and 2.12 ± 0.01 g, respectively.

    Click to Show/Hide
Response regulation Microcystin-LR induces intestinal injury. Lesion morphological features (vacuolization, deformation and dilation of the endoplasmic reticulum [ER], absence of mitochondrial cristae in mid-intestine), up-regulated mRNA expressions of ER stress (eukaryotic translation initiation factor 2-alpha kinase 3, endoplasmic reticulum to nucleus signaling 1, activating transcription factor [ATF] 6, ATF4, DNA damage-inducible transcript 3), iron accumulation, and down-regulated activity of glutathione peroxidase (GPx) and glutathione (GSH) content were all typical characteristics of ferroptosis in intestinal tissue following MC-LR exposure.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [7]
Responsed Disease Anti-tuberculosis drug-induced liver injury [ICD-11: NB91]
Responsed Drug Rifampicin Investigative
Responsed Regulator Heat shock cognate 71 kDa protein (HSPA8) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model Hep-G2 cells Hepatoblastoma Homo sapiens CVCL_0027
AML12 cells Normal Mus musculus CVCL_0140
In Vivo Model
Male C57BL/6 mice (8-10 weeks old and weighing 20-25 g) were purchased from Hunan Experimental Animal Center (Changsha, China). All animals were housed in a 12/12h light/dark cycle and given free access to water and food. All experimental procedures were conducted in accordance with the institutional guidelines for animal care. After a minimum of 7 days of acclimation, the mice were randomly divided into five groups. DILI model group was given rifampicin (350 mg/kg) that was dissolved in saline by gavage daily for 14 days, and the same volume of saline was given as the control. Meanwhile, the other three groups were given the same volume of rifampicin to establish drug liver injury model, and treated with different compounds in the second week as follows: ferrostatin 1 (0.6 mg/kg) was injected into the tail vein every day after 2 h of rifampicin administration; geldanamycin (HSP90 inhibitor) (0.75 mg/kg) was intraperitoneally injected 2 h after rifampicin daily; 3-methyladenine (autophagy inhibitor) (15 mg/kg) was injected into tail vein 2 h after rifampicin every day. After 2 weeks of administration, serum was collected, and then mice were sacrificed by cervical dislocation. After that, liver tissue was taken for different treatments and used for reserve.

    Click to Show/Hide
Response regulation Inhibition of HSPA8 by rifampicin contributes to ferroptosis via enhancing autophagy. The present study highlights the crucial roles of the HSPA8 and autophagy in ferroptotic cell death driving by rifampicin, particularly illumines multiple promising regulatory nodes for therapeutic interventions in diseases involving anti-tuberculosis drug-induced liver injury (AT-DILI).
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Responsed Disease Acetaminophen-induced liver injury [ICD-11: DB95.Z]
Responsed Drug VBIT-12 Investigative
Responsed Regulator Sphingomyelin phosphodiesterase (SMPD1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model mPHs (Mouse primary hepatocytes)
In Vivo Model
Male C57 BL /6J mice (6-8 weeks; 18-22 g each) were procured from the Experimental Animal Centre of Shanghai SLAC. All mice were fed in laboratory in vivo facilities with ad libitum food and water, within a temperature-/humidity-regulated environment (22 ± 1 ; Rh. = 65 ± 5%), adopting a 12-h circadian cycling process. Mice fasted overnight for 12 h and were randomly divided into different groups.

    Click to Show/Hide
Response regulation The relative amounts of CER and CL in the liver tissues of VBIT-12- or UAMC3203-treated APAP-injured mice markedly increased compared to those of APAP-injured mice. Smpd1 was the CER synthesis gene that was most significantly upregulated by UAMC3203 and VBIT-12. In summary, protecting mitochondria via inhibiting VDAC1 oligomerization alleviates ferroptosis in acetaminophen-induced acute liver injury.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [22]
Responsed Disease DEHP-induced liver injury [ICD-11: NB91]
Responsed Drug Diethylhexylphthalate Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Glutathione metabolism hsa00480
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
In Vivo Model
Adult male and female fish were randomly assigned to 6 aquariums and acclimated to laboratory conditions for one week before DEHP exposure. There are 6 aquariums, where 3 aquariums were used for DEHP exposure as the treatment groups and 3 for control. Each aquarium was filled with 12 L of artificial seawater and 30 females and 30 males were assigned. Food residues and excrement were removed, and artificial water was renewed every day. The exposure groups were exposed to 10 ug/L DEHP, obtained by diluting the stock solution, and the control groups were treated with DMSO as a solvent control, in which the DMSO content did not exceed 0.01% (v/v).

    Click to Show/Hide
Response regulation Ferroptosis occurred in response to di (2-ethylhexyl) phthalate (DEHP) exposure, which resulted in DEHP-induced liver injury, an enrichment of the ferroptosis pathway along with iron overload, an increase in malondialdehyde (MDA) and lipid peroxidation (LPO) content, and a decrease in glutathione (GSH) levels.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [23]
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Drug Tyrosylalanine Investigative
Pathway Response Fatty acid metabolism hsa01212
Apoptosis hsa04210
Ferroptosis hsa04216
Necroptosis hsa04217
Cell Process Cell ferroptosis
Cell apoptosis
Cell pyroptosis
In Vitro Model mLTs (Mouse liver tissues)
In Vivo Model
Male C57BL/6 mice (7 weeks old) were purchased from Koatech Co. (Animal Breeding Center, Pyongtaek, Korea). Animals were kept on a 12 h light/dark cycle in a specific pathogen-free area with food and water freely available in the animal facility for 1 week before the experiment. All experimental animals were randomly separated into five groups as follows: Saline, LPS (1 g/kg) + D-GalN (400 mg/kg), LPS/D-GalN + YA (10 mg/kg), LPS/D-GalN + YA (50 mg/kg), and LPS/D-GalN + silymarin (25 mg/kg). YA and silymarin were pre-administered for 10 days before LPS/D-GalN by oral gavage. LPS/D-GalN was injected intraperitoneally. Blood and tissues were collected 6 h after LPS/D-GalN injection.

    Click to Show/Hide
Response regulation Tyrosine-alanine (YA) had a hepatoprotective effect by reducing inflammation, apoptosis, ferroptosis, and pyroptosis in the LPS/D-GalN-injected ALF mouse model. YA can be used as a functional peptide for the prevention of acute liver injury.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Regulator NADPH--cytochrome P450 reductase (POR) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
HeLa cells Endocervical adenocarcinoma Homo sapiens CVCL_0030
mEFs (Mouse embryonic fibroblasts)
HEK-293T cells Normal Homo sapiens CVCL_0063
OVCAR-8 cells High grade ovarian serous adenocarcinoma Homo sapiens CVCL_1629
In Vivo Model
Female nu/nu mice aged 4-5 weeks were obtained from Charles River. Luciferase expresing-OVCAR-8 cells were harvested by trypsinization. Subsequently, cells were washed three times with cold PBS and suspended in a 1:1 mixture of PBS and Matrigel (Corning). Each mouse was inoculated subcutaneously with 5 x 106 cells. When tumor volume reached approximately 50 mm3, mice were randomly divided into indicated groups. 20 mg PACMA31 per kg body weight (10% DMSO, 30% PEG-4000, 60% Saline); 40 mg regorafenib per kg body weight (Saline); or 20 mg PACMA31 plus 40 mg regorafenib per kg body weight daily. PACMA31 was intraperitoneally injected and regorafenib was orally administered.

    Click to Show/Hide
Response regulation Genetic knockout of POR and CYB5R1 decreases cellular hydrogen peroxide generation, preventing lipid peroxidation and ferroptosis. Moreover, POR knockdown in mice confers protective effects during acute liver injury (ALI) caused from ferroptosis.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Regulator NADH-cytochrome b5 reductase 1 (CYB5R1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
HeLa cells Endocervical adenocarcinoma Homo sapiens CVCL_0030
mEFs (Mouse embryonic fibroblasts)
HEK-293T cells Normal Homo sapiens CVCL_0063
OVCAR-8 cells High grade ovarian serous adenocarcinoma Homo sapiens CVCL_1629
In Vivo Model
Female nu/nu mice aged 4-5 weeks were obtained from Charles River. Luciferase expresing-OVCAR-8 cells were harvested by trypsinization. Subsequently, cells were washed three times with cold PBS and suspended in a 1:1 mixture of PBS and Matrigel (Corning). Each mouse was inoculated subcutaneously with 5 x 106 cells. When tumor volume reached approximately 50 mm3, mice were randomly divided into indicated groups. 20 mg PACMA31 per kg body weight (10% DMSO, 30% PEG-4000, 60% Saline); 40 mg regorafenib per kg body weight (Saline); or 20 mg PACMA31 plus 40 mg regorafenib per kg body weight daily. PACMA31 was intraperitoneally injected and regorafenib was orally administered.

    Click to Show/Hide
Response regulation Genetic knockout of POR and CYB5R1 decreases cellular hydrogen peroxide generation, preventing lipid peroxidation and ferroptosis. Moreover, POR knockdown in mice confers protective effects during acute liver injury (ALI) caused from ferroptosis.
Phospholipid hydroperoxide glutathione peroxidase (GPX4)
In total 6 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Target for Ferroptosis Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Kaempferol Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
Male BALB/c mice (8-week-old, 20-22 g) were purchased from Guangdong Medical Laboratory Animal Center (Guangzhou, China). The experimental animals were fed adaptively for one week in the Experimental Animal Center of Guangdong Pharmaceutical University (Guangzhou, China). Feeding conditions were set at 26 , humidity 65% and a lightdark cycle for 12 hours. All animal experiments were performed following the Guide for the Care and Use of Laboratory Animals, and the procedures were approved by the Research Ethical Committee of Guangdong Pharmaceutical University (gdpulacspf2020007).

    Click to Show/Hide
Response regulation Kaempferol (KA) activated the Nrf2 pathway and upregulated Gpx4 in mouse livers and L02 cells to inhibit ferroptosis induced by APAP. Finally, molecular docking indicated the potential interaction of KA with Keap1. Taken together, KA ameliorated oxidative stress and ferroptosis-mediated acetaminophen-induced liver injury by activating Nrf2 signaling.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Target for Ferroptosis Suppressor
Responsed Disease DEHP-induced liver injury [ICD-11: NB91]
Responsed Drug Apigenin Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model AML12 cells Normal Mus musculus CVCL_0140
Response regulation DEHP caused oxidative stress and increased the Fe2+ content, finally resulting in ferroptosis in AML12 cells. Apigenin restrained the toxicity of DEHP and antagonized DEHP-induced ferroptosis in AML12 cells. The protective effects of APG on DEHP-induced liver injury were achieved by activating GPX4 and suppressing intracellular iron accumulation.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Target for Ferroptosis Suppressor
Responsed Disease CCl4-induced iver injury [ICD-11: NB91]
Responsed Drug Bicyclol Investigative
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model L-02 cells Endocervical adenocarcinoma Homo sapiens CVCL_6926
In Vivo Model
The mice were treated with intraperitoneal administration (i.p.) of oil (control group) or a mixture of CCl4 (50%) and oil (50%) at a dosage of 2 ml/kg body weight. In the bicyclol-treated group, mice accepted administration of 200 mg/kg (using 0.5% carboxymethyl cellulose as solvent) by gavage three times a day 1 h before CCl4 exposure, while other groups accepted vehicles of the equal volume. Fer-1 was prepared in DMSO (5 mg/kg), andi.p. injected into mice once 1 h before CCl4 exposure. The dosage of bicyclol was consistent with our previous work. The mice were then sacrificed to collect liver and serum samples after 24 or 48 h.

    Click to Show/Hide
Response regulation Bicyclol exerted its hepatoprotection by preventing the aforesaid ferroptotic process. Furthermore, bicyclol alleviated erastin-induced cellular inviability, destruction, and lipid peroxidation in vitro. Knockdown of GPX4 diminished these protective activities against perturbations associated with ferroptosis in L-O2 hepatocytes. Additionally, Nrf2 silencing drastically reduced GPX4 levels, and further impeded the medicinal effects of bicyclol. In summary, positively regulating Nrf2-GPX4 axis by bicyclol can prevent ferroptosis in CCl4-induced acute liver injury in mice.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Target for Ferroptosis Suppressor
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Drug Disulfiram Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
HEK-293T cells Normal Homo sapiens CVCL_0063
786-O cells Renal cell carcinoma Homo sapiens CVCL_1051
769-P cells Renal cell carcinom Homo sapiens CVCL_1050
SK-HEP-1 cells Liver and intrahepatic bile duct epithelial neoplasm Homo sapiens CVCL_0525
HCCLM3 cells Adult hepatocellular carcinoma Homo sapiens CVCL_6832
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
MDA231-LM2-4175 cells Breast adenocarcinoma Homo sapiens CVCL_5998
In Vivo Model
C57BL/6J male mice aged 8 weeks were purchased from Charles River Laboratories International, Inc., and housed in a specific pathogen-free animal facility. DMSO or DSF (21 mg/kg) was injected intraperitoneally into mice for 0.5 h, followed by ConA injection via the tail vein at 15 mg/kg. Mice were sacrificed at 24 h post ConA injection. Liver and blood samples were collected at this time point for H&E staining, IHC staining, and measurement of AST/ALT (Dian Diagnostics Co., Ltd).

    Click to Show/Hide
Response regulation Disulfiram (DSF) is conjugated to multiple cysteine residues in GPX4 and disrupts GPX4 interaction with HSC70, an adaptor protein for chaperone mediated autophagy, thus preventing GPX4 degradation induced by erastin. In addition, DSF ameliorates concanavalin A induced acute liver injury by suppressing ferroptosis in a mouse model.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [13]
Target for Ferroptosis Suppressor
Responsed Disease Liver Oxidative Damage [ICD-11: NB91]
Responsed Drug Epigallocatechin Gallate Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
In Vivo Model
All mice were randomly divided into a 2 x 2 factorial arrangement, fed diets containing 40 mg/kg or 5000 mg/kg FeSO4 (the basis of the diet was AIN-93), and gavaged with PBS or 50 mg EGCG/kg body weight per day, respectively. The experiment lasted for 6 weeks, including a 1-week adaptation and a 3-week EGCG gavage; then, all mice were euthanized.

    Click to Show/Hide
Response regulation Epigallocatechin-3-Gallate (EGCG) supplementation alleviated the liver oxidative damage caused by iron overload by inhibiting ferroptosis. EGCG addition increased NRF2 and GPX4 expression and elevated antioxidant capacity in iron overload mice. EGCG administration attenuates iron metabolism disorders by upregulating FTH/FTL expression. Through these two mechanisms, EGCG can effectively inhibit iron overload-induced ferroptosis.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [14]
Target for Ferroptosis Suppressor
Responsed Disease Pirarubicin-induced hepatotoxicity [ICD-11: NB91]
Responsed Drug Schisandrin B Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model rPHs (Rat primary hepatocytes)
In Vivo Model
A total of 40 male SD rats (180~200 g, 6~8 weeks) were purchased from the CMU experimental animal center. The rats were randomly divided into four groups: control (CON) group (normal diet rats were injected with equal volume of normal saline through caudal vein once a week, n = 10), SchB group (SchB diet rats, 50 mg/kg/day, were injected with equal volume of normal saline through caudal vein once a week, n = 10), THP group (normal diet rats were injected with 3 mg/kg/day THP through caudal vein once a week, n = 10), and SchB+THP group (SchB diet rats, 50 mg/kg/day, were injected with 3 mg/kg/day THP through caudal vein once a week, n = 10). CON and THP rats were fed an AIN-76A feed (12.4% fat, 68.8% carbohydrate, and 18.8% protein). SchB and SchB+THP rats were fed an SchB feed (approximately 0.5 SchB was added into AIN-76A feed). After conversion, 0.5 SchB in feed = 50 mg/kg in rats.

    Click to Show/Hide
Response regulation Schisandrin B (SchB) increased the levels of SOD, GSH, GSH-px, CAT, and T-AOC, decreased the level of MDA, and inhibited the abnormal oxidative stress in the liver. And SchB as a natural molecule depends on reducing the level of oxidative stress, thereby inhibiting lipid peroxidation, ferroptosis, and apoptosis. The expression of NRF2, GPX4, SOD2, and Bcl-2/Bax decreased, while the expression of NOX2/4 and cleaved caspase-3 increased in pirarubicin-treated hepatocytes. However, the above changes were significantly reversed after SchB or Fer-1 treatment. SchB has obvious protective effect on pirarubicin-induced hepatotoxicity.
Nuclear receptor coactivator 4 (NCOA4)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Driver
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Quercetin Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model Hep-G2 cells Hepatoblastoma Homo sapiens CVCL_0027
In Vivo Model
Six-week-old male C57BL/6J mice (18-20 g) were obtained from Zhejiang Vital River Laboratory (Zhejiang, China). 32 mice were divided randomly into 4 groups: Saline group (CONT), 25 mg/kg/day ACR group (ACR), 25 mg/kg/day ACR with a low dose of 25 mg/kg/day QCT group (ACR + QCT (L)), and 25 mg/kg/day ACR with a high dose of 50 mg/kg/day QCT group (ACR + QCT (H)), 8 animals in each group.

    Click to Show/Hide
Response regulation Quercetin (QCT) specifically reacted with autophagic cargo receptor NCOA4, blocked the degradation of iron storage protein FTH1, and eventually downregulated the intracellular iron levels and the consequent ferroptosis. Collectively, our results presented a unique approach to alleviate ACR-induced liver injury by targeting ferroptosis with QCT.
Ferritin light chain (FTL)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [13]
Target for Ferroptosis Suppressor
Responsed Disease Liver Oxidative Damage [ICD-11: NB91]
Responsed Drug Epigallocatechin Gallate Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
In Vivo Model
All mice were randomly divided into a 2 x 2 factorial arrangement, fed diets containing 40 mg/kg or 5000 mg/kg FeSO4 (the basis of the diet was AIN-93), and gavaged with PBS or 50 mg EGCG/kg body weight per day, respectively. The experiment lasted for 6 weeks, including a 1-week adaptation and a 3-week EGCG gavage; then, all mice were euthanized.

    Click to Show/Hide
Response regulation Epigallocatechin-3-Gallate (EGCG) supplementation alleviated the liver oxidative damage caused by iron overload by inhibiting ferroptosis. EGCG addition increased NRF2 and GPX4 expression and elevated antioxidant capacity in iron overload mice. EGCG administration attenuates iron metabolism disorders by upregulating FTH/FTL expression. Through these two mechanisms, EGCG can effectively inhibit iron overload-induced ferroptosis.
Ferritin heavy chain (FTH1)
In total 2 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [13]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Liver Oxidative Damage [ICD-11: NB91]
Responsed Drug Epigallocatechin Gallate Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
In Vivo Model
All mice were randomly divided into a 2 x 2 factorial arrangement, fed diets containing 40 mg/kg or 5000 mg/kg FeSO4 (the basis of the diet was AIN-93), and gavaged with PBS or 50 mg EGCG/kg body weight per day, respectively. The experiment lasted for 6 weeks, including a 1-week adaptation and a 3-week EGCG gavage; then, all mice were euthanized.

    Click to Show/Hide
Response regulation Epigallocatechin-3-Gallate (EGCG) supplementation alleviated the liver oxidative damage caused by iron overload by inhibiting ferroptosis. EGCG addition increased NRF2 and GPX4 expression and elevated antioxidant capacity in iron overload mice. EGCG administration attenuates iron metabolism disorders by upregulating FTH/FTL expression. Through these two mechanisms, EGCG can effectively inhibit iron overload-induced ferroptosis.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Drug Quercetin Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model Hep-G2 cells Hepatoblastoma Homo sapiens CVCL_0027
In Vivo Model
Six-week-old male C57BL/6J mice (18-20 g) were obtained from Zhejiang Vital River Laboratory (Zhejiang, China). 32 mice were divided randomly into 4 groups: Saline group (CONT), 25 mg/kg/day ACR group (ACR), 25 mg/kg/day ACR with a low dose of 25 mg/kg/day QCT group (ACR + QCT (L)), and 25 mg/kg/day ACR with a high dose of 50 mg/kg/day QCT group (ACR + QCT (H)), 8 animals in each group.

    Click to Show/Hide
Response regulation Quercetin (QCT) specifically reacted with autophagic cargo receptor NCOA4, blocked the degradation of iron storage protein FTH1, and eventually downregulated the intracellular iron levels and the consequent ferroptosis. Collectively, our results presented a unique approach to alleviate ACR-induced liver injury by targeting ferroptosis with QCT.
Cystine/glutamate transporter (SLC7A11)
In total 4 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [19]
Target for Ferroptosis Suppressor
Responsed Disease DEHP-induced splenic toxicity [ICD-11: NB91]
Responsed Drug Lycopene Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model mSCs (Mouse splenocytes)
In Vivo Model
Three-week-old specific pathogen-free ICR (Institute of Cancer Research) male mice (weights of 18-22 g) were provided by Liaoning Changsheng Biotech Co. Ltd. The mice were housed under conditions at 22 ± 2 with 35-65% humidity and a light/dark cycle of 12 h/12 h in the cage. The animals were quarantined for a week before formal experiments, then randomly divided into seven groups: vehicle control group (Vcon), control group (Con), 5 mg/kg BW/d Lyc group (Lyc), 500 and 1000 mg/kg BW/d DEHP group (D5 and D10, respectively), DEHP + Lyc group (DL5 and DL10, respectively) (n = 20). The animals were exposed to DEHP via oral gavage, which lasted for 28 d, and then sacrificed after being anesthetized.

    Click to Show/Hide
Response regulation DEHP disrupted the GSH metabolism via the xc/ GPX4 antioxidant system and, subsequently, caused the ferroptotic cell death, but Lycopene (Lyc) could effectively mitigate DEHP-induced damage to the antioxidant system. These findings indicated that Lyc may be an effective strategy for the prevention of DEHP-induced splenic toxicity via the regulation of ferroptosis.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [20]
Target for Ferroptosis Suppressor
Responsed Disease Liver injury [ICD-11: NB91]
Responsed Regulator gga-miR-129-3p (miRNA) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model LMH cells Hepatoma Gallus gallus CVCL_2580
LMH cells Hepatoma Gallus gallus CVCL_2580
Response regulation miR-129-3p affected ferroptosis under Se deficiency conditions through the SLC7A11 pathway. Our research provides a new perspective for the mechanism of Se deficiency on the liver damage.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [21]
Target for Ferroptosis Suppressor
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Regulator CD44 antigen (CD44) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Ubiquitin mediated proteolysis hsa04120
Cell Process Cell ferroptosis
In Vitro Model mPHs (Mouse primary hepatocytes)
In Vivo Model
ALI induction was performed in 6-8-week-old age-matched C57BL/6J male mice (n = 10-12 per group) by intraperitoneal injection of 3 mL/kg CCl4 in coconut oil. Control and negative control mice were injected with PBS and coconut oil, respectively. At 6 h after injection of CCl4, mice were divided into three groups: CCl4 group, injected with 100 uL PBS (supplemented with 2% mouse serum) through a tail vein; CCl4 + MSC group, injected with 5 x 105 MSCs suspended in 100 uL PBS (supplemented with 2% mouse serum) through a tail vein; CCl4 + Fer-1 group, intraperitoneally injected with ferrostatin-1 (Fer-1, a ferroptosis inhibitor, 2.5 umol/kg body weight). Erastin, intraperitoneal injection of erastin (a ferroptosis inducer, 30 mg/kg body weight) twice every other day, and then the mice were divided into two groups (n = 10-12 per group): Erastin group, injected with 100 uL PBS (supplemented with 2% mouse serum) through a tail vein; Erastin + MSC group, injected with 5 x 105 MSCs suspended in 100 uL PBS (supplemented with 2% mouse serum) through the tail vein.

    Click to Show/Hide
Response regulation MSC-Exo protected against CCl4-induced acute liver injury (ALI) through inhibiting hepatocyte ferroptosis via restoring the SLC7A11 protein level. Additionally, the exosome-induced recovery of SLC7A11 protein was accompanied by upregulations of CD44 and OTUB1.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [21]
Target for Ferroptosis Suppressor
Responsed Disease Acute liver injury [ICD-11: NB91]
Responsed Regulator Ubiquitin thioesterase OTUB1 (OTUB1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Ubiquitin mediated proteolysis hsa04120
Cell Process Cell ferroptosis
In Vitro Model mPHs (Mouse primary hepatocytes)
In Vivo Model
ALI induction was performed in 6-8-week-old age-matched C57BL/6J male mice (n = 10-12 per group) by intraperitoneal injection of 3 mL/kg CCl4 in coconut oil. Control and negative control mice were injected with PBS and coconut oil, respectively. At 6 h after injection of CCl4, mice were divided into three groups: CCl4 group, injected with 100 uL PBS (supplemented with 2% mouse serum) through a tail vein; CCl4 + MSC group, injected with 5 x 105 MSCs suspended in 100 uL PBS (supplemented with 2% mouse serum) through a tail vein; CCl4 + Fer-1 group, intraperitoneally injected with ferrostatin-1 (Fer-1, a ferroptosis inhibitor, 2.5 umol/kg body weight). Erastin, intraperitoneal injection of erastin (a ferroptosis inducer, 30 mg/kg body weight) twice every other day, and then the mice were divided into two groups (n = 10-12 per group): Erastin group, injected with 100 uL PBS (supplemented with 2% mouse serum) through a tail vein; Erastin + MSC group, injected with 5 x 105 MSCs suspended in 100 uL PBS (supplemented with 2% mouse serum) through the tail vein.

    Click to Show/Hide
Response regulation MSC-Exo protected against CCl4-induced acute liver injury (ALI) through inhibiting hepatocyte ferroptosis via restoring the SLC7A11 protein level. Additionally, the exosome-induced recovery of SLC7A11 protein was accompanied by upregulations of CD44 and OTUB1.
References
Ref 1 Ulinastatin protects against acetaminophen-induced liver injury by alleviating ferroptosis via the SIRT1/NRF2/HO-1 pathway. Am J Transl Res. 2021 Jun 15;13(6):6031-6042. eCollection 2021.
Ref 2 Exosomes Derived from Baicalin-Pretreated Mesenchymal Stem Cells Alleviate Hepatocyte Ferroptosis after Acute Liver Injury via the Keap1-NRF2 Pathway. Oxid Med Cell Longev. 2022 Jul 21;2022:8287227. doi: 10.1155/2022/8287227. eCollection 2022.
Ref 3 (+)-Clausenamide protects against drug-induced liver injury by inhibiting hepatocyte ferroptosis. Cell Death Dis. 2020 Sep 19;11(9):781. doi: 10.1038/s41419-020-02961-5.
Ref 4 Xiaojianzhong decoction attenuates gastric mucosal injury by activating the p62/Keap1/Nrf2 signaling pathway to inhibit ferroptosis. Biomed Pharmacother. 2022 Nov;155:113631. doi: 10.1016/j.biopha.2022.113631. Epub 2022 Sep 16.
Ref 5 Protective effect of sestrin2 against iron overload and ferroptosis-induced liver injury. Toxicol Appl Pharmacol. 2019 Sep 15;379:114665. doi: 10.1016/j.taap.2019.114665. Epub 2019 Jul 16.
Ref 6 Microcystin-LR induces ferroptosis in intestine of common carp (Cyprinus carpio). Ecotoxicol Environ Saf. 2021 Oct 15;223:112610. doi: 10.1016/j.ecoenv.2021.112610. Epub 2021 Aug 5.
Ref 7 Inhibition of HSPA8 by rifampicin contributes to ferroptosis via enhancing autophagy. Liver Int. 2022 Dec;42(12):2889-2899. doi: 10.1111/liv.15459. Epub 2022 Oct 26.
Ref 8 Protecting mitochondria via inhibiting VDAC1 oligomerization alleviates ferroptosis in acetaminophen-induced acute liver injury. Cell Biol Toxicol. 2022 Jun;38(3):505-530. doi: 10.1007/s10565-021-09624-x. Epub 2021 Aug 17.
Ref 9 Kaempferol prevents acetaminophen-induced liver injury by suppressing hepatocyte ferroptosis via Nrf2 pathway activation. Food Funct. 2023 Feb 21;14(4):1884-1896. doi: 10.1039/d2fo02716j.
Ref 10 Apigenin ameliorates di(2-ethylhexyl) phthalate-induced ferroptosis: The activation of glutathione peroxidase 4 and suppression of iron intake. Food Chem Toxicol. 2022 Jun;164:113089. doi: 10.1016/j.fct.2022.113089. Epub 2022 Apr 30.
Ref 11 Regulating Nrf2-GPx4 axis by bicyclol can prevent ferroptosis in carbon tetrachloride-induced acute liver injury in mice. Cell Death Discov. 2022 Sep 7;8(1):380. doi: 10.1038/s41420-022-01173-4.
Ref 12 A Class of Disulfide Compounds Suppresses Ferroptosis by Stabilizing GPX4. ACS Chem Biol. 2022 Dec 16;17(12):3389-3406. doi: 10.1021/acschembio.2c00445. Epub 2022 Nov 29.
Ref 13 Epigallocatechin-3-Gallate Alleviates Liver Oxidative Damage Caused by Iron Overload in Mice through Inhibiting Ferroptosis. Nutrients. 2023 Apr 21;15(8):1993. doi: 10.3390/nu15081993.
Ref 14 Schisandrin B Diet Inhibits Oxidative Stress to Reduce Ferroptosis and Lipid Peroxidation to Prevent Pirarubicin-Induced Hepatotoxicity. Biomed Res Int. 2022 Jul 18;2022:5623555. doi: 10.1155/2022/5623555. eCollection 2022.
Ref 15 Quercetin Alleviates Acrylamide-Induced Liver Injury by Inhibiting Autophagy-Dependent Ferroptosis. J Agric Food Chem. 2023 May 17;71(19):7427-7439. doi: 10.1021/acs.jafc.3c01378. Epub 2023 May 3.
Ref 16 Abietic acid inhibits acetaminophen-induced liver injury by alleviating inflammation and ferroptosis through regulating Nrf2/HO-1 axis. Int Immunopharmacol. 2023 May;118:110029. doi: 10.1016/j.intimp.2023.110029. Epub 2023 Mar 22.
Ref 17 Astaxanthin Activated the Nrf2/HO-1 Pathway to Enhance Autophagy and Inhibit Ferroptosis, Ameliorating Acetaminophen-Induced Liver Injury. ACS Appl Mater Interfaces. 2022 Sep 28;14(38):42887-42903. doi: 10.1021/acsami.2c10506. Epub 2022 Sep 12.
Ref 18 Inhibitor of apoptosis-stimulating protein of p53 inhibits ferroptosis and alleviates intestinal ischemia/reperfusion-induced acute lung injury. Cell Death Differ. 2020 Sep;27(9):2635-2650. doi: 10.1038/s41418-020-0528-x. Epub 2020 Mar 18.
Ref 19 Potential Role of Lycopene in the Inhibition of Di(2-ethylhexyl) Phthalate-Induced Ferroptosis in Spleen Via Modulation of Iron Ion Homeostasis. ACS Pharmacol Transl Sci. 2021 Jan 27;4(1):386-395. doi: 10.1021/acsptsci.1c00001. eCollection 2021 Feb 12.
Ref 20 MiR-129-3p regulates ferroptosis in the liver of Selenium-deficient broilers by targeting SLC7A11. Poult Sci. 2023 Jan;102(1):102271. doi: 10.1016/j.psj.2022.102271. Epub 2022 Oct 27.
Ref 21 Mesenchymal stem cells protect against ferroptosis via exosome-mediated stabilization of SLC7A11 in acute liver injury. Cell Death Dis. 2022 Mar 26;13(3):271. doi: 10.1038/s41419-022-04708-w.
Ref 22 Acute exposure of di(2-ethylhexyl) phthalate (DEHP) induces immune signal regulation and ferroptosis in oryzias melastigma. Chemosphere. 2021 Feb;265:129053. doi: 10.1016/j.chemosphere.2020.129053. Epub 2020 Nov 23.
Ref 23 Oyster-Derived Tyr-Ala (YA) Peptide Prevents Lipopolysaccharide/D-Galactosamine-Induced Acute Liver Failure by Suppressing Inflammatory, Apoptotic, Ferroptotic, and Pyroptotic Signals. Mar Drugs. 2021 Oct 28;19(11):614. doi: 10.3390/md19110614.
Ref 24 Membrane Damage during Ferroptosis Is Caused by Oxidation of Phospholipids Catalyzed by the Oxidoreductases POR and CYB5R1. Mol Cell. 2021 Jan 21;81(2):355-369.e10. doi: 10.1016/j.molcel.2020.11.024. Epub 2020 Dec 14.