Ferroptosis Regulator Information
General Information of the Ferroptosis Regulator (ID: REG10520)
Full List of the Ferroptosis Target of This Regulator and Corresponding Disease/Drug Response(s)
TP53
can regulate the following target(s), and cause disease/drug response(s). You can browse detail information of target(s) or disease/drug response(s).
Browse Target
Browse Disease
Browse Drug
Phospholipid hydroperoxide glutathione peroxidase (GPX4) [Suppressor]
In total 5 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis Target of This Regulator | [1] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Oral squamous cell carcinoma | ICD-11: 2B6E | |||
Responsed Drug | Quisinostat | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Necroptosis | hsa04217 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell apoptosis | |||||
Cell pyroptosis | |||||
In Vitro Model |
CAL-27 cells | Tongue adenosquamous carcinom | Homo sapiens | CVCL_1107 | |
Tca8113 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6851 | ||
In Vivo Model |
Adult male athymic BALB/c nude mice (20-22 g of 5-week-old mice) were housed in a controlled environment at 23 ± 2 and 40%-70% humidity under a 12 h dark/light cycle with free access to irradiated food and sterile water. A suspension of 6 x 106/100 uL TCA-8113 cells was inoculated subcutaneously into the hind flank region of each nude mouse. The average tumor volume in nude mice reached 100 mm3, and mice were randomly divided into three groups. Quisinostat was formulated in normal saline and administered at 3 and 10 mg/kg/day byintraperitoneal injection. Control mice were given equal volume saline intraperitoneally. The tumor volume and the bodyweight of mice were monitored every three days.
Click to Show/Hide
|
||||
Response regulation | Quisinostat could increase the apoptosis rate in the tumor tissues of nude mice. Up-regulation of the expression of p53 and down-regulated expression of GPX4 in cell lines were observed by immunofluorescent staining, and the expression locations of p53 and GPX4 proteins in TSCC cells were observed. Quisinostat may be a potential drug for the treatment of tongue squamous cell carcinoma. | ||||
Experiment 2 Reporting the Ferroptosis Target of This Regulator | [2] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Status epilepticus | ICD-11: 8A66 | |||
Responsed Drug | Apigenin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
SH-SY5Y cells | Neuroblastoma | Homo sapiens | CVCL_0019 | |
In Vivo Model |
5-weeks-old kainate (KA)-induced BALB/c nude mice, a widely used epilepsy mouse model, were performed with intraperitoneal (i.p.) injection of KA (6 mg/kg). Pre-treatment 21 with antioxidant apigenin (60 mg/Kg, 2 days) or post-treatment with apigenin (60 mg/Kg, 1 day), mice were injected with KA (6 mg/kg) via intraperitoneal (i.p.) injection, and then HCP (0.5 mg/Kg) were injected by intravenous (i.v.) injection. In vivo and Ex vivo fluorescence images of relative ClO levels in mice brains 5, 15, 30, 45, and 60 min post injection of HCP were further performed by using the IVIS Spectrum imaging system (Nanjing University) with an excitation filter of 430 nm and the collection wavelength range is from 500-600 nm.
Click to Show/Hide
|
||||
Response regulation | Apigenin can efficiently reduce the expression of intracellular MPO and increase the levels of GPX4 and SIRT1, thereby conferring neuroprotection through regulation of kainic acid (KA)-induced ferroptosis. And the level of Ac-p53 inside the brains treated with apigenin was down-regulated, suggesting that the p53-mediated ferroptosis pathway might be blocked. Overall, apigenin was screened and confirmed as an efficient lead compound for epilepsy prevention and treatment. | ||||
Experiment 3 Reporting the Ferroptosis Target of This Regulator | [30] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Colon cancer | ICD-11: 2B90 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
HeLa cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0030 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
HIEC-6 cells | Normal | Homo sapiens | CVCL_6C21 | ||
Response regulation | RRM1 increases the instability of p53 by regulating the physical interaction of p53 with the ubiquitinating enzyme MDM2 and the deubiquitinating enzyme USP11, subsequently suppressing p21 (CDKN1A) and GPX4 in colon carcinoma cells, thereby promoting the accumulation of lipid peroxidation and occurrence of radiation-induced ferroptosis. | ||||
Experiment 4 Reporting the Ferroptosis Target of This Regulator | [31] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Colorectal cancer | ICD-11: 2B91 | |||
Pathway Response | Ferroptosis | hsa04216 | |||
MAPK signaling pathway | hsa04010 | ||||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
SW480 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0546 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
In Vivo Model |
Six 4-week-old male BALB/c nude mice were ordered from the Shanghai Laboratory Animal Center (Shanghai SLAC Laboratory Animal Co., Ltd., China). A total of 5 x 106 TIPE+/+ SW480 cells were suspended in 100 uL of PBS and subcutaneously injected into the right axilla flank of each nude mouse, and the same amount of vector SW480 cells was into the left. At 2 weeks after inoculation, the xenograft tumor size was measured using Vernier calipers every 2 days.
Click to Show/Hide
|
||||
Response regulation | MiR-539 can bind to and regulate the expression of TIPE, and miR-539 activates SAPK/JNK to downregulate the expression of glutathione peroxidase 4 (GPX4) and promote ferroptosis. In addition, SAPK/JNK is the upstream molecule of p53. MiR-539 is a new therapeutic target for colorectal cancer (CRC) patients. | ||||
Experiment 5 Reporting the Ferroptosis Target of This Regulator | [32] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Pancreatic cancer | ICD-11: 2C10 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
BxPC-3 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0186 | |
PANC-1 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0480 | ||
Response regulation | Cold induction promotes the process of ferroptosis by inducing the expression of CIRBP and then regulating key factors such as p53 and GPX4[. In addition, cold induction significantly inhibited the proliferation of pancreatic cancer cells and induced cell apoptosis, but after the addition of ferroptosis inhibitor, cell proliferation and apoptosis did not change significantly. | ||||
Diamine acetyltransferase 1 (SAT1) [Driver]
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis Target of This Regulator | [3] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Neurotoxicity | ICD-11: NE61 | |||
Responsed Drug | Sevoflurane | Approved | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hBCs (Brain cells) | ||||
In Vivo Model |
Pregnant rats were placed in a dedicated plastic chamber with ambient gas at a flow rate of 2L/min. Fer-1 solubilized in saline and 1% dimethyl sulfoxide (DMSO) and PD146176 (a specific 15LOX inhibitor) dissolved in corn oil containing 1% DMSO were administered intraperitoneally to rats at a dose of 5 mg/kg 1 h before each exposure, respectively. Similarly, 0.5 mg/kg Ku55933 (an ATM inhibitor), which is diluted in saline containing 1% DMSO, was intraperitoneally administered 2 h previously.
Click to Show/Hide
|
||||
Response regulation | Sevoflurane could enhance 15LO2-PEBP1 interaction and activate ATM and its downstream P53/SAT1 pathway, which might be attributed to excessive p-ATM nuclear translocation, indicating a potential therapeutic target for ameliorating sevoflurane-induced neurotoxicity. | ||||
Cystine/glutamate transporter (SLC7A11) [Driver; Suppressor]
In total 10 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis Target of This Regulator | [4] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma | ICD-11: 2A00 | |||
Responsed Drug | Pseudolaric acid B | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
U-87MG cells | Glioblastoma | Homo sapiens | CVCL_0022 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
SHG-44 cells | Astrocytoma | Homo sapiens | CVCL_6728 | ||
In Vivo Model |
Twenty athymic BALB/c nude mice (aged 4 weeks, weight 20-22 g, from Shanghai laboratory animal Center, Shanghai, China) were housed in a specific pathogen-free environment. A total of 1 x 106 logarithmically growing C6 cells in 100 uL of PBS were subcutaneously injected into the right flank of each mouse. Therapeutic experiments were started when the tumor reached about 150 mm3 after about 7 days. The mice were allocated to receive intraperitoneal injections of vehicle (control group, n = 5/group), PAB at the dosage of 10 mg/kg body weight (n = 10/group) and 20 mg/kg body weight (n = 10/group) in the same volume 50 uL once a days for 8 times.
Click to Show/Hide
|
||||
Response regulation | Pseudolaric acid B (PAB) improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT (SLC7A11) pathway, which further exacerbated accumulation of H2O2and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment. | ||||
Experiment 2 Reporting the Ferroptosis Target of This Regulator | [5] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | T-cell lymphoma | ICD-11: 2B01 | |||
Responsed Drug | Kayadiol | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
YT cells | Natural killer cell lymphoblastic leukemia | Homo sapiens | CVCL_1797 | |
hPBLs (Human peripheral blood lymphocytes) | |||||
Response regulation | Kayadiol decreased the expression of SLC7A11 and GPX4, the negative regulatory proteins for ferroptosis. And p53 was the key mediator of kayadiol-induced ferroptosis by SLC7A11/GPX4 axis through p53 knockout experiments. Kayadiol can serve as an effective alternative in the treatment of NK/T cell lymphoma. | ||||
Experiment 3 Reporting the Ferroptosis Target of This Regulator | [6] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Osteosarcoma | ICD-11: 2B51 | |||
Responsed Drug | Bavachin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
MG-63 cells | Osteosarcoma | Homo sapiens | CVCL_0426 | |
HOS cells | Osteosarcoma | Homo sapiens | CVCL_0312 | ||
Response regulation | Bavachin could induce Osteosarcoma cell ferroptosis. Furthermore, bavachin elevated intracellular ferrous iron levels by increasing TFRC and DMT1 expression and decreasing FTH and FTL expressions. Bavachin also reduced SLC7A11 and GPX4 expression and promoted ROS and MDA accumulation by downregulating p-STAT3 to upregulate P53 expression. | ||||
Experiment 4 Reporting the Ferroptosis Target of This Regulator | [7] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Gastric cancer | ICD-11: 2B72 | |||
Responsed Drug | Tanshinone IIA | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
BGC-823 cells | Gastric carcinoma | Homo sapiens | CVCL_3360 | |
NCI-N87 cells | Gastric tubular adenocarcinoma | Homo sapiens | CVCL_1603 | ||
In Vivo Model |
All mice were housed under a setting of 12-h light/dark cycle at 22 ± 1, 55% humidity and fed with water and food provided at regular time. During the entire maintenance period, all mice were permitted free cage activity without joint immobilization. The initial body weights of the mice were between 20 and 23 grams. After subcutaneous injection of 2 x 106 BGC-823 gastric cancer cells into the back of NOD-SCID mice, the mice were treated with or without Tan IIA (50 mg/kg) or Tan IIA in combination with Fer-1 (50 mg/kg). Tan IIA was diluted in DMSO:Methanol:Hydroxypropyl-b-cydodextrin (HP-b-CD) = 1:1:1. Fer-1 was also dissolved in DMSO:Methanol:HP-b-CD. Seven days after BGC-823 gastric cancer cells injection, intraperitoneal injection with Tan IIA was carried out every other day followed by killing at day 22 of tumor cell inoculation. All mice were killed by dislocation of the cervical vertebrae. Before killing, the tumor volume was measured every 3 days. All experiments were carried out using six mice each group in three independent experiments of a time-dependent manner with three time points.
Click to Show/Hide
|
||||
Response regulation | Tanshinone IIA increased lipid peroxidation and up-regulated Ptgs2 and Chac1 expression, two markers of ferroptosis. In addition, Tan IIA also up-regulated p53 expression and down-regulated xCT (SLC7A11) expression. Therefore, Tan IIA could suppress the proliferation of gastric cancer via inducing p53 upregulation-mediated ferroptosis. | ||||
Experiment 5 Reporting the Ferroptosis Target of This Regulator | [8] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Melanoma | ICD-11: 2C30 | |||
Responsed Drug | Gambogenic Acid | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell adhesion molecules | hsa04514 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model |
A-375 cells | Amelanotic melanoma | Homo sapiens | CVCL_0132 | |
A2058 cells | Amelanotic melanoma | Homo sapiens | CVCL_1059 | ||
Response regulation | Gambogenic acid (GNA) significantly inhibited the invasion, migration and EMT in melanoma cells, and these cells exhibited small mitochondrial wrinkling (an important feature of ferroptosis). GNA upregulated the expression of p53, solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) in the model cells, contributing to the mechanisms underlying GNA-induced ferroptosis. | ||||
Experiment 6 Reporting the Ferroptosis Target of This Regulator | [9] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Parkinson disease | ICD-11: 8A00 | |||
Responsed Drug | Cyperquat | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
PC12 cells | Adrenal gland pheochromocytoma | Rattus norvegicus | CVCL_0481 | |
Response regulation | As a classic drug employed inin vitromodels of Parkinson's disease, 1-methyl-4-phenylpyridinium (MPP) can induce senescence in PC12 cells. The expression of the ferroptosis-related proteins ASCL4 was upregulated and FTH1 was downregulated, which promoted accumulation of lipid peroxides and eventually led to ferroptosis. By rescuing MPP-induced ferroptosis, cell senescence could be inhibited, and its molecular mechanism was related to a p53/SLC7A11/GPX4 signaling pathway. | ||||
Experiment 7 Reporting the Ferroptosis Target of This Regulator | [10] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Status epilepticus | ICD-11: 8A66 | |||
Responsed Drug | Seratrodast | Discontinued in Phase 3 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
HT22 cells | Normal | Mus musculus | CVCL_0321 | |
In Vivo Model |
Drugs were dissolved in vehicle (0.1% DMSO + 20% PEG 300 + 0.5% CMC-Na + ddH2O). Mice in Control and PTZ groups were administered for five days with an equivalent volume of vehicle. PTZ-induced seizure model was done for the subsequent 1 h after the last administration of drugs. We performed a preliminary doseresponse trial, the dose of 60 mg/kg was established as being sufficient to trigger seizures with lower mortality and chosen as the optimal dose. One mouse in PTZ group was dead due to a severe seizure. At the end of the experiment, the mice were anesthetized or euthanized. For histopathological studies, the mice were anesthetized and intracardially perfused with 0.9% saline, followed by 0.4% paraformaldehyde for fixation of the brain. For immunoblot analysis, the hippocampus was rapidly isolated.
Click to Show/Hide
|
||||
Response regulation | Seratrodast could reduce lipid ROS production, regulate the system xc-/glutathione (GSH)/glutathione peroxidase 4 (GPX4) axis, and inhibit JNK (MAPK8) phosphorylation and p53 expression. JNK can directly or indirectly modulate the expression and activation of p53, which could regulate ferroptosis through inhibition of SLC7A11 transcription. Seratrodast increased the latency of seizures and reduced seizure duration in pentylenetetrazole-induced seizures in Epilepsy. | ||||
Experiment 8 Reporting the Ferroptosis Target of This Regulator | [11] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Intracerebral hemorrhage | ICD-11: 8B00 | |||
Responsed Drug | Isorhynchophylline | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
p53 signaling pathway | hsa04115 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
HT22 cells | Normal | Mus musculus | CVCL_0321 | |
In Vivo Model |
Adult male Sprague-Dawley rats (SD rats, weighing 250-300 g) aged 11-12 weeks were purchased from SLAC Laboratory Animal Co., Ltd. (Shanghai, China). All 96 rats were randomly divided into four groups of 24 rats each: Sham group, Sham + IRN (30 mg/Kg) group, ICH group, and ICH + IRN (30 mg/Kg) group. The rats in sham group were injected with PBS solution, and the Sham + IRN (30 mg/Kg) group was received an equal amount of 30 mg/Kg IRN solution (intra-peritoneal injection) after the sham operation. After ICH, the rats in ICH group were injected with PBS solution, and the ICH + IRN (30 mg/Kg) group was received an equal amount of 30 mg/Kg IRN solution (intra-peritoneal injection).
Click to Show/Hide
|
||||
Response regulation | Isorhynchophylline (IRN) decreased ferroptosis and lipid ROS level, upregulated the expression of miR-122-5p and SLC7A11 mRNA, and inhibited TP53 expression. In conclusion, IRN protects neurocyte from intracerebral hemorrhage (ICH)-induced ferroptosis via miR-122-5p/TP53/SLC7A11 pathway, which may provide a potential therapeutic mechanism for ICH. | ||||
Experiment 9 Reporting the Ferroptosis Target of This Regulator | [12] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Sarcopenia | ICD-11: FB32 | |||
Responsed Drug | Ferric citrate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Glutathione metabolism | hsa00480 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
C2C12 cells | Normal | Mus musculus | CVCL_0188 | |
In Vivo Model |
The 8-week- and 40-week-old male SAMP8 mice were purchased from the model animal research center of Zhishan Institute of Healthcare Research Co., Ltd. (Beijing, China). All the mice were kept in an SPF grade animal facility at 24 with a relative humidity of 50%-60%, and in a light/dark cycle of 12 h/12 h.
Click to Show/Hide
|
||||
Response regulation | Ferric citrate induced ferroptosis in C2C12 cells, as well as impaired their differentiation from myoblasts to myotubes. Iron overload upregulated the expression of P53, which subsequently repressed the protein level of Slc7a11 (solute carrier family 7, member 11), a known ferroptosis-related gene. Targeting iron accumulation and ferroptosis might be a therapeutic strategy for treating sarcopenia. | ||||
Experiment 10 Reporting the Ferroptosis Target of This Regulator | [34] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Lung injury | ICD-11: NB32 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
THP-1 cells | Childhood acute monocytic leukemia | Homo sapiens | CVCL_0006 | |
HBE1 cells | Normal | Homo sapiens | CVCL_0287 | ||
In Vivo Model |
For the models of CS and LPS exposure, mice were anesthetized and intratracheally instilled with CS suspensions (3 mg/50 ul) or LPS (1 mg/kg). For the models of CS + Ferr-1/DFO, mice were intraperitoneally injected with Ferr-1 (1.25 umol/kg) or intranasal instilled with DFO (10 mg/kg) for 7 consecutive days after CS instillation. For the models of LPS + Ferr-1/DFO, mice were pretreated with Ferr-1 or DFO for 2 consecutive days and then intratracheally instilled with LPS. Mice were sacrificed 24 h after LPS instillation. For the X-ray exposure model, mice were exposed to ionizing radiation (IR) at 20 Gy, which was delivered at the dose rate of 2 Gy/min and a source skin distance of 51 cm by an X-ray generator (Model X-RAD320iX; Precision X-Ray, Inc., North Branford, CT, USA), and sacrificed 3 days after radiation.
Click to Show/Hide
|
||||
Response regulation | STAT6 negatively regulates ferroptosis through competitively binding with CBP, which inhibits P53 acetylation and transcriptionally restores SLC7A11 expression. Finally, pulmonary-specific STAT6 overexpression decreased the ferroptosis and attenuated CS and LPS induced acute lung injury. | ||||
Unspecific Target [Unspecific Target]
In total 30 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis Target of This Regulator | [13] | ||||
Responsed Disease | HIV Infection | ICD-11: 1C60 | |||
Responsed Drug | Methamphetamine | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
BV-2 cells | Normal | Mus musculus | CVCL_0182 | |
Response regulation | Methamphetamine (METH) and HIV-1 lead to oxidative stress and their combined effect increases the risk of HIV-associated neurocognitive disorder (HAND), which may be related to the synergistic ferroptotic impairment in microglia. We found that METH and HIV-1 Tat reduced the expression of ferroptotic protein GPX4 and the cell viability and enhanced the expression of P53 and the level of ferrous iron, while the above indices were significantly improved with pretreatment of ferrostatin-1. | ||||
Experiment 2 Reporting the Ferroptosis Target of This Regulator | [14] | ||||
Responsed Disease | Colon cancer | ICD-11: 2B90 | |||
Responsed Drug | Albiziabioside A | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
MCF-7 cells | Breast carcinoma | Homo sapiens | CVCL_0031 | |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | ||
hBMECs (Human brain microvascular endothelial cells) | |||||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
HaCaT cells | Normal | Homo sapiens | CVCL_0038 | ||
HCT-8 cells | Ileocecal adenocarcinoma | Homo sapiens | CVCL_2478 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
CCD-841CoN cells | Normal | Homo sapiens | CVCL_2871 | ||
BEAS-2B cells | Normal | Homo sapiens | CVCL_0168 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
In Vivo Model |
A total of 1 x 107 HCT116 cells were subcutaneously inoculated into the right flank of BALB/c mice. When tumor reached 70-100 mm3 (10 days after implant), mice were divided into five groups of eight animals at random. The groups with D13 were administered intravenously 20 mg/kg and 10 mg/kg. The positive control group was treated with AlbA(20 mg/kg and 10 mg/kg) through intravenous injection. The negative control group received 0.9% normal saline through intravenous injection.
Click to Show/Hide
|
||||
Response regulation | The study synthesized a series of Albiziabioside A derivatives and evaluated the antitumor activity both in vitro and in vivo. Compound D13 could induce apoptosis and ferroptosis through the mitochondrial pathway as a p53 activator. In addition, compound D13 significantly suppressed tumorigenesis without inducing toxicity in normal organs in vivo. The antitumor efficacy of D13 was further verified in colon cancer xenograft mouse models. | ||||
Experiment 3 Reporting the Ferroptosis Target of This Regulator | [15] | ||||
Responsed Disease | Colon cancer | ICD-11: 2B90 | |||
Responsed Drug | Ginkgo biflavones | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
HT29 cells | Colon cancer | Mus musculus | CVCL_A8EZ | ||
RKO cells | Colon carcinoma | Homo sapiens | CVCL_0504 | ||
LoVo cells | Colon adenocarcinoma | Homo sapiens | CVCL_0399 | ||
SW620 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0547 | ||
In Vivo Model |
HCT-116 cells (5 x 106) were injected into the flanks of 6-week-old male BALB/c nude mice to generate xenografts. The mice were randomly divided into four groups (n = 7 per group), and treatment was started at 96 h postinjection. The mice received an intraperitoneal injection (i.p.) of 0.9% saline solution containing 5% dimethyl sulfoxide (DMSO) (vehicle for blank control) and ginkgetin (10 mg/kg) once a day, respectively, and 5-FU (30 mg/kg) was used as the positive control (i.p., once every 3 days, alternately).
Click to Show/Hide
|
||||
Response regulation | Ginkgo biflavones can increase the expression level of p53 by inhibiting the expression of MDM2 protein and induce cell death independent of p53 transcriptional activity in vitro. And we provide evidence that ginkgetin strengthened the antitumor effect of fluorouracil (5-FU) in the HCT-116 colon cancer xenograft model. | ||||
Experiment 4 Reporting the Ferroptosis Target of This Regulator | [16] | ||||
Responsed Disease | Colorectal cancer | ICD-11: 2B91 | |||
Responsed Drug | BEBT-908 | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
PI3K-Akt signaling pathway | hsa04151 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
MC-38 cells | Colon adenocarcinoma | Homo sapiens | CVCL_B288 | |
In Vivo Model |
About 2 x 105 MC38 tumor cells were inoculated subcutaneously into C57BL/6 mice. A total of 100 mg/kg BEBT-908 was intravenously injected every other day for four times initiating from day 4 after tumor cell inoculation. Tumors were harvested on day 12 after inoculation, weighted, mechanically minced and incubated with 50 ug/mL DNase I (Sigma) and 2 mg/mL collagenase P (Sigma) for 20 minutes at 37.
Click to Show/Hide
|
||||
Response regulation | Treatment with BEBT-908 promoted ferroptotic cell death of cancer cells by hyperacetylating p53 and facilitating the expression of ferroptotic signaling. The dual PI3K/HDAC inhibitor BEBT-908 elicits potent antitumor responses, effectively inducing immunogenic ferroptosis of colorectal cancer cells and potentiating cancer immunotherapy. | ||||
Experiment 5 Reporting the Ferroptosis Target of This Regulator | [17] | ||||
Responsed Disease | Pancreatic cancer | ICD-11: 2C10 | |||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
Panc02 cells | Pancreatic ductal adenocarcinoma | Mus musculus | CVCL_D627 | |
PANC-1 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0480 | ||
In Vivo Model |
Six to eight-week-old female C57BL/6 mice were purchased from the Experimental Animal Center of Military Medical Sciences (Beijing, China). C57BL/6 mice were anesthetized and the tail of the pancreas was exposed. Panc 02 cells were resuspended in PBS at a concentration of 1 x 106 cells/0.1 ml and 50 ul cells were injected into the tail of the pancreas. Tumor-bearing mice were randomly divided into two groups (3 days after implantation). The control group was intraperitoneally injected 200 ul PBS daily for 10 days, and the DHA group was intraperitoneally injected with 100 mg/kg DHA daily for 10 days. The pancreatic tumors and spleens of the mice were collected for subsequent analysis.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin has anti-tumor effect in pancreatic cancer cells in vitro and in vivo. DHA treatment induced ferroptosis by increasing P53 and AOLX12 expression. | ||||
Experiment 6 Reporting the Ferroptosis Target of This Regulator | [18] | ||||
Responsed Disease | Lung cancer | ICD-11: 2C25 | |||
Responsed Drug | Levobupivacaine | Approved | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model |
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | |
A427 cells | Lung carcinoma | Homo sapiens | CVCL_1055 | ||
In Vivo Model |
Balb/c nude mice (n= 5, 4-week-old, male) were applied to detect the impact of levobupivacaine on tumor growth. Mice subcutaneously injected with 1 x 107 A549 cells were treated with levobupivacaine (40 umol/Kg) or equal volume saline. The tumor volume was remarked every 5 days and finished at 30 days after injection, followed by the analysis of volume (length (width/2)2) and weight.
Click to Show/Hide
|
||||
Response regulation | Levobupivacaine could inhibit the proliferation and induce the apoptosis of non-small cell lung cancer (NSCLC) cells. Levobupivacaine was able to attenuate the invasion and migration in the cells. The treatment of levobupivacaine remarkably increased the levels of ROS, iron, and Fe2+ in NSCLC cells. Mechanically, levobupivacaine up-regulated the expression of p53 and induced ferroptosis by regulating p53 in NSCLC cells. | ||||
Experiment 7 Reporting the Ferroptosis Target of This Regulator | [19] | ||||
Responsed Disease | Ovarian cancer | ICD-11: 2C73 | |||
Responsed Drug | Apatinib | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model |
A2780 cells | Ovarian endometrioid adenocarcinoma | Homo sapiens | CVCL_0134 | |
OVCAR-3 cells | Ovarian serous adenocarcinoma | Homo sapiens | CVCL_0465 | ||
Response regulation | Apatinib combined with olaparib-induced ferroptosis via a p53-dependent manner in ovarian cancer. Further studies showed that apatinib combined with olaparib-induced ferroptosis by inhibiting the expression of Nrf2 and autophagy, thereby inhibiting the expression of GPX4. The Nrf2 activator RTA408 and the autophagy activator rapamycin rescued the combination drug-induced ferroptosis. | ||||
Experiment 8 Reporting the Ferroptosis Target of This Regulator | [19] | ||||
Responsed Disease | Ovarian cancer | ICD-11: 2C73 | |||
Responsed Drug | Olaparib | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
In Vitro Model |
A2780 cells | Ovarian endometrioid adenocarcinoma | Homo sapiens | CVCL_0134 | |
OVCAR-3 cells | Ovarian serous adenocarcinoma | Homo sapiens | CVCL_0465 | ||
Response regulation | Apatinib combined with olaparib-induced ferroptosis via a p53-dependent manner in ovarian cancer. Further studies showed that apatinib combined with olaparib-induced ferroptosis by inhibiting the expression of Nrf2 and autophagy, thereby inhibiting the expression of GPX4. The Nrf2 activator RTA408 and the autophagy activator rapamycin rescued the combination drug-induced ferroptosis. | ||||
Experiment 9 Reporting the Ferroptosis Target of This Regulator | [20] | ||||
Responsed Disease | Prostate cancer | ICD-11: 2C82 | |||
Responsed Drug | Flubendazole | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell apoptosis | |||||
In Vitro Model |
PC-3 cells | Prostate carcinoma | Homo sapiens | CVCL_0035 | |
DU145 cells | Prostate carcinoma | Homo sapiens | CVCL_0105 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
RWPE-1 cells | Normal | Homo sapiens | CVCL_3791 | ||
In Vivo Model |
24 nude mice (3-4 weeks) were acquired from the experimental animal center of southern medical university (GuangZhou, China) and kept under specific pathogen-free conditions. 4 x 106 PC3 cells were implanted subcutaneously into the right armpit regions of each nude mouse. When the tumors volume reached approximately 40 mm3, mice were randomly divided into two groups to receive flubendazole (10 mg/kg, once daily), 5-fluorouracil (30 mg/kg, once daily), their combination, and vehicle control by intraperitoneal injection. After 20 days of treatment, all mice were sacrificed and tumor weight and tumor volume were immediately measured, respectively.
Click to Show/Hide
|
||||
Response regulation | Flubendazole is a novel P53 inducer which exerts anti-proliferation and pro-apoptosis effects in castration-resistant prostate cancer (CRPC) through hindering the cell cycle and activating the ferroptosis, and indicates that a novel utilization of flubendazole in neoadjuvant chemotherapy of CRPC. | ||||
Experiment 10 Reporting the Ferroptosis Target of This Regulator | [21] | ||||
Responsed Disease | Hereditary Leiomyomatosis | ICD-11: 2C90 | |||
Responsed Drug | Artesunate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
Caki-1 cells | Clear cell renal cell carcinoma | Homo sapiens | CVCL_0234 | |
786-O cells | Renal cell carcinoma | Homo sapiens | CVCL_1051 | ||
KTCTL-26 cells | Clear cell renal cell carcinoma | Homo sapiens | CVCL_5872 | ||
A-498 cells | Renal cell carcinoma | Homo sapiens | CVCL_1056 | ||
Response regulation | Artesunate (ART) significantly increased cytotoxicity and inhibited proliferation and clonogenic growth in both parental and sunitinib-resistant renal cell carcinoma (RCC) cells. P53 exclusively appeared in the KTCTL-26 cells, indicating that p53 might be predictive for ART-dependent ferroptosis. Thus, ART may hold promise for treating selected patients with advanced and even therapy-resistant RCC. | ||||
Experiment 11 Reporting the Ferroptosis Target of This Regulator | [22] | ||||
Responsed Disease | Adrenocortical carcinoma | ICD-11: 2D11 | |||
Responsed Drug | Romidepsin | Investigative | |||
Pathway Response | Cell adhesion molecules | hsa04514 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
SW13 cells | Adrenal cortex carcinoma | Homo sapiens | CVCL_0542 | |
Response regulation | HDAC inhibitor Romidepsin converted SW13+ cells also had reduced mRNA expression of the mitochondrial ROS detoxifier superoxide dismutase 2 (SOD2), and the tumor suppressor p53. HDAC inhibitor treatment synergistically increased adrenocortical carcinoma cell death following induction of ferroptosis. | ||||
Experiment 12 Reporting the Ferroptosis Target of This Regulator | [23] | ||||
Responsed Disease | Parkinson disease | ICD-11: 8A00 | |||
Responsed Drug | Clioquinol | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
PI3K-Akt signaling pathway | hsa04151 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
SK-N-SH cells | Neuroblastoma | Homo sapiens | CVCL_0531 | |
In Vivo Model |
In total, twelve healthy adult rhesus monkeys (Macaca mulatta lasiotis, aged 4-5 years, and weighed 3.5-5 kg at the start of the study) were obtained from Sichuan Primed Biological Technology Co., Ltd. Monkeys were randomly divided into two groups: normal (control) group (n = 3) and MPTP group (n = 9). Monkeys from MPTP group were administered with MPTP by intramuscular injection daily at the beginning of the study, and then the MPTP dose was gradually added to 0.5 mg/kg at the end of the experiment. Monkeys from control group were injected with saline instead, and the other conditions were the same with MPTP group.
Click to Show/Hide
|
||||
Response regulation | Ferroptosis was probably involved in the pathogenesis of parkinson's disease (PD). Clioquinol (CQ) can decrease the excessive iron in the SN to normal level and directly protect DA neurons against oxidative stress probably by activating the AKT/mTOR survival pathway and blocking p53-medicated cell death. | ||||
Experiment 13 Reporting the Ferroptosis Target of This Regulator | [24] | ||||
Responsed Disease | Parkinson disease | ICD-11: 8A00 | |||
Responsed Drug | Ferric ammonium citrate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
MES23.5 cells | Normal | Mus musculus | CVCL_J351 | |
In Vivo Model |
Human a-synuclein (a-Syn) A53T overexpressiontransgenic mice(B6; C3-Tg (Prnp-SNCA*A53T) 83Vle/J) were originally obtained in breeding pairs from the Jackson Laboratory (004479) to generate a stable breeding colony. Animals were raised according to SPF level, kept at constant temperature (20 ± 2) , constant humidity (50 ± 10%), day and night cycle light (12-12 h), with free access to food and water.
Click to Show/Hide
|
||||
Response regulation | Ferroptosis firstly occurred in a relatively low concentration of ferric ammonium citrate (FAC)-treated group, and then apoptosis appeared in response to the increased iron doses. This was also confirmed in vivo in parkinson's disease transgenic mice and the underlying mechanism might be associated with the p53 signaling pathway, but not MAPK signaling pathway. | ||||
Experiment 14 Reporting the Ferroptosis Target of This Regulator | [25] | ||||
Responsed Disease | Cardiomyopathy | ICD-11: BC43 | |||
Responsed Drug | Berberine | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
All animal experiment protocols were implemented in accordance with the National Institutes of Health (NIH) guidelines, and the procedures were approved by the Animal Ethics Committee of Southwest University. C57BL/6J male mice, 8-10 weeks old, weighing 20 ± 2 g, were used in this study. Mice were housed under standard conditions at 22-24 with a 12 h light/12 h darkness cycle and free access to food and tap water. Thirty-six mice were randomly divided into six groups: control (N = 8), IMA group (50 mg/kg) (N = 8), Low-Ber (20 mg/kg) + IMA group (N = 8), Medium-Ber (40 mg kg1) + IMA group (N = 8), High-Ber (80 mg/kg) + IMA group (N = 8), and Fer-1 (1 mg/kg) + IMA group (N = 8). IMA was given intraperitoneally for 14 days. Ber was given orally 2 h before IMA treatment and Fer-1 was given intraperitoneally 2 h before IMA treatment.
Click to Show/Hide
|
||||
Response regulation | Berberine downregulated the expression of transferrin receptor (TfR) and P53 and upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), ferritin heavy chain-1 (FTH1), and glutathione peroxidase 4 (GPX4) in H9c2 cells and mice. The present data indicated that Ber has the potential to protect against IMA-induced cardiotoxicity, partlyviainhibiting Nrf2-dependent ferroptosis. | ||||
Experiment 15 Reporting the Ferroptosis Target of This Regulator | [26] | ||||
Responsed Disease | Osteonecrosis | ICD-11: FB81 | |||
Responsed Drug | Zoledronic acid | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
RAW 264.7 cells | Leukemia | Mus musculus | CVCL_0493 | |
Response regulation | FBXO9 was downregulated in Zoledronic acid-treated osteoclast and promoted osteoclasts ferroptosis by inhibiting FBXO9-mediated p53 ubiquitination and degradation. The study provided a possible theoretical target for the clinical treatment of Bisphosphonates (BPs)-related osteonecrosis of jaw (BRONJ). | ||||
Experiment 16 Reporting the Ferroptosis Target of This Regulator | [27] | ||||
Responsed Disease | Lung injury | ICD-11: NB32 | |||
Responsed Drug | Lipopolysaccharide | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
MLE-12 cells | Normal | Mus musculus | CVCL_3751 | |
Response regulation | Silence of MLK3 (MAP3K11) alleviated Lipopolysaccharide (LPS)-induced lung epithelial cell injury by inhibiting p53-mediated ferroptosis, suggesting that MLK3 may be a potential target to prevent acute lung injury. | ||||
Experiment 17 Reporting the Ferroptosis Target of This Regulator | [35] | ||||
Responsed Disease | Acute myeloid leukaemia | ICD-11: 2A60 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model |
hBMSCs (Bone marrow stromal cells) | ||||
K-562 cells | Chronic myelogenous leukemia | Homo sapiens | CVCL_0004 | ||
THP-1 cells | Childhood acute monocytic leukemia | Homo sapiens | CVCL_0006 | ||
HL-60 cells | Adult acute myeloid leukemia | Homo sapiens | CVCL_0002 | ||
MOLM-13 cells | Adult acute myeloid leukemia | Homo sapiens | CVCL_2119 | ||
In Vivo Model |
All BALB/C male nude mice (6 weeks old), purchased from Beijing Weitong lihua Experimental Animal Technology Co., Ltd, were maintained in pathogen-free facilities. The K562 or HL-60 cells (3 x 106, 200 ul) transfected with circKDM4C, vector, si-NC, or si-circKDM4C were subcutaneously injected into the right flank of nude mice (9 mice each group) for tumorigenesis. The maximum (Length) and minimum (Width) length of the tumors were measured.
Click to Show/Hide
|
||||
Response regulation | The overexpression of circKDM4C in acute myeloid leukemia (AML) cell lines inhibited the cell proliferation, migration, invasion, and promoted ferroptosis. The expression of circKDM4C and hsa-let-7b-5p are negatively correlated, while circKDM4C and p53 are positively correlated to AML patients. Moreover, circKDM4C induces ferroptosis by sponging hsa-let-7b-5p which upregulates the expression of P53. | ||||
Experiment 18 Reporting the Ferroptosis Target of This Regulator | [36] | ||||
Responsed Disease | Osteosarcoma | ICD-11: 2B51 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
U2OS cells | Osteosarcoma | Homo sapiens | CVCL_0042 | |
IMR-90 cells | Normal | Homo sapiens | CVCL_0347 | ||
Response regulation | SOCS1 impacts the pattern of secreted products in cells with active p53 and is required for the expression of a selective set of p53 target genes including those involved in ferroptosis. SOCS1 can use several mechanisms to activate p53, including promotion of serine-15 phosphorylation by ATM/ATR kinases and inhibition of the p53 repressor KAP1 in osteosarcoma cell lines. | ||||
Experiment 19 Reporting the Ferroptosis Target of This Regulator | [37] | ||||
Responsed Disease | Colon cancer | ICD-11: 2B90 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CT26 cells | Colon adenocarcinoma | Mus musculus | CVCL_7254 | |
4T1 cells | Mammary carcinoma | Mus musculus | CVCL_0125 | ||
In Vivo Model |
BALB/c and C57BL/6 mice were obtained from the Jackson Laboratory (Bar Harbor, ME). To establish subcutaneous tumor models, CT26 cells (2 x 105 cells/mouse) were injected into the right flanks of BALB/c mice. For experimental lung metastasis models, colon carcinoma CT26, mesothelioma AB1 (2 x 105 cells/mouse) and mammary carcinoma 4T1 (2 x 104 cells/mouse) were injected into BALB/c mice. Tumor-bearing mice were treated with vehicle PEG300 (Sigma-Aldrich) and NC06 (dissolved in PEG300), respectively, every two days for 3-5 times by Intraperitoneal injection.
Click to Show/Hide
|
||||
Response regulation | ASAH2 is overexpressed in MDSCs in human colon cancer patients. ASAH2 protects MDSCs from ferroptosis through destabilizing p53 protein to suppress the p53 pathway in MDSCs in the tumor microenvironment. | ||||
Experiment 20 Reporting the Ferroptosis Target of This Regulator | [38] | ||||
Responsed Disease | Colon cancer | ICD-11: 2B90 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Hippo signaling pathway | hsa04390 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
SW620 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0547 | ||
Response regulation | CYGB significantly increased the sensitivity of cancer cells to RSL3- and erastin-induced ferroptotic cell death. Collectively, a novel tumour suppressor role of CYGB through p53-YAP1 axis in regulating ferroptosis and suggested a potential therapeutic approach for colon cancer. | ||||
Experiment 21 Reporting the Ferroptosis Target of This Regulator | [39] | ||||
Responsed Disease | Hepatocellular carcinoma | ICD-11: 2C12 | |||
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 22 Reporting the Ferroptosis Target of This Regulator | [40] | ||||
Responsed Disease | Lung cancer | ICD-11: 2C25 | |||
Pathway Response | Cell adhesion molecules | hsa04514 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
NCI-H460 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0459 | ||
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
NCI-H358 cells | Minimally invasive lung adenocarcinoma | Homo sapiens | CVCL_1559 | ||
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
In Vivo Model |
The 4-wk-old female nude mice used in this study were purchased from Hunan SJA Laboratory Animal Co., Ltd (Changsha). Then, 2 x 106 GINS4 WT or KO A549 cells were injected s.c. into nude mice without matrigel. Tumor size was measured every 2 d with a caliper, and volume of tumor was calculated with the formula: L x W2 x 0.5, for L represents the longest diameter and W means the shortest diameter. Then, 2 x 106 p53 WT and KO A549 cells with shCon or shGINS4 overexpressing were injected s.c. into nude mice without matrigel. When tumors reached 60 to 100 mm3, p53 WT with shGINS4 mice were treated with ferrostatin-1 (S7243, Selleckchem) (20 mg/kg) in 2% DMSO, 50% PEG300, 5% Tween80, and 43% water by daily intraperitoneal injection. Tumors were measured three times a week. Mice were sacrificed and tumors were collected finally. Tumor tissue was made into a single-cell suspension for lipid ROS assay using Tumor Dissociation Kit (Miltenyi Biotec).
Click to Show/Hide
|
||||
Response regulation | GINS4 is a potential oncogene in lung adenocarcinoma (LUAD) that functions to destabilize p53 and then inhibits ferroptosis, providing a potential therapeutic target for LUAD. | ||||
Experiment 23 Reporting the Ferroptosis Target of This Regulator | [41] | ||||
Responsed Disease | Lung cancer | ICD-11: 2C25 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | |
SPC-A1 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6955 | ||
NCI-H522 cells | Non-small cell lung carcinoma | Homo sapiens | CVCL_1567 | ||
HBE1 cells | Normal | Homo sapiens | CVCL_0287 | ||
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
MRC-5 cells | Normal | Homo sapiens | CVCL_0440 | ||
NCI-H358 cells | Minimally invasive lung adenocarcinoma | Homo sapiens | CVCL_1559 | ||
PC-9 cells | Lung adenocarcinoma | Homo sapiens | CVCL_B260 | ||
95C cells | Lung giant cell carcinoma | Homo sapiens | CVCL_7109 | ||
95D cells | Lung giant cell carcinoma | Homo sapiens | CVCL_7110 | ||
In Vivo Model |
SCID mice (Hunan SJA Laboratory Animal Co. Ltd.) were injected with the indicated cells in the mammary fat pad (10 mice/group). Injected mice were imaged from both the dorsal and ventral sides every three days.
Click to Show/Hide
|
||||
Response regulation | P53RRA bound Ras GTPase-activating protein-binding protein 1 (G3BP1) using nucleotides 1 and 871 of P53RRA and the RRM interaction domain of G3BP1 (aa 177-466). The cytosolic P53RRA-G3BP1 interaction displaced p53 from a G3BP1 complex, resulting in greater p53 retention in the nucleus, which led to cell-cycle arrest, apoptosis, and ferroptosis in lung cancer cell lines. | ||||
Experiment 24 Reporting the Ferroptosis Target of This Regulator | [42] | ||||
Responsed Disease | Cerebral ischemia | ICD-11: 8B10 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
Wild-type SD rats were kept in the Animal Experiment Center of Southeast University. Experimental rats were divided into 4 groups (n = 6 per group). The method of establishing the I/R model was provided in supplementary material. Then, we covered the ligation with gel. In order to fully cover the infarcted area of the heart, we chose to inject about 300 uL of mimics + Gel at 23 mm below the left atrial appendage (about the ligation). In order to prevent excessive irradiation of tissue burns, we selected each irradiation for 2 min to control the body surface temperature for a total of 10 min of irradiation.
Click to Show/Hide
|
||||
Response regulation | The mir-196c-3p mimic (mimics) and photothermal nanoparticles (BTN) were co-encapsulated in an injectable Gel (mimics + Gel/BTN) with NIR-II light-triggered release. Consequently, declined ferroptosis in cardiomyocytes and improved cardiac function, survival rate in rats was achieved through the controlled release of Gel/BTN mimics in ischemia-reperfusion (I/R) model to simultaneously inhibit ferroptosis hub genes NOX4, P53, and LOX expression. | ||||
Experiment 25 Reporting the Ferroptosis Target of This Regulator | [43] | ||||
Responsed Disease | Cerebral ischaemic stroke | ICD-11: 8B11 | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
rRBMECs (Rat brain microvascular endothelial cells) | ||||
In Vivo Model |
Sprague Dawley (SD) rats (n = 60) aged three weeks were purchased from the Experimental Animal Center of Xiangya Hospital of Central South University. All rats were bred in a specific pathogen-free environment in 12-h lightdark cycle and fed with rodent diet and water. All rats were anaesthetized with inhaling isoflurane (2%, CAS NO. 64181101, Lunan Pharmaceutical Co., LTD. Shandong, China) and sacrificed by cervical dislocation. The whole brain was removed after opening the cranial cavity.
Click to Show/Hide
|
||||
Response regulation | OGD combined with hyperglycemic reperfusion promoted Meg3 expression and there was positive correlation between Meg3 and p53 expression in RBMVECs. Subsequently, p53 inhibited the activity of GPX4 by binding with its promoter. The Meg3- p53 signaling pathway mediated the ferroptosis of RBMVECs upon injury induced by OGD combined with hyperglycemic reperfusion and Meg3 has been considered as an important mediator in regulating diabetic brain ischemic injury. | ||||
Experiment 26 Reporting the Ferroptosis Target of This Regulator | [44] | ||||
Responsed Disease | Cerebral ischaemic stroke | ICD-11: 8B11 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
PC12 cells | Adrenal gland pheochromocytoma | Rattus norvegicus | CVCL_0481 | |
Response regulation | PVT1 regulates ferroptosis through miR-214-mediated p53 and TFR1. The discovery of PVT1 and miR-214 as potential targets for I/R also implies that PVT1 and miR-214 play critical roles in ferroptosis, shedding new light on the mechanism of ferroptosis in acute ischemic stroke. | ||||
Experiment 27 Reporting the Ferroptosis Target of This Regulator | [45] | ||||
Responsed Disease | Chronic heart failure | ICD-11: BD1Z | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Necroptosis | hsa04217 | ||||
NF-kappa B signaling pathway | hsa04064 | ||||
Cell Process | Cell ferroptosis | ||||
Cell pyroptosis | |||||
In Vitro Model |
rHTs (Rat hippocampal tissues) | ||||
In Vivo Model |
Male wild-type (WT) C57BL/6J mice aged 8 weeks were obtained from the Experimental Animal Center, Guangzhou University of Chinese Medicine. Sham and TAC mice received corresponding isotype i.p. injections. TAC + AAVMLK3- mice were generated by intravenous (i.v.) injection of adeno-associated viral vector-MLK3 vector (AAVMLK3-) (GenePharma, Shanghai, China) 14 and 21 days before TAC surgery. Sham + AAVNC and TAC + AAVNC mice received AAVNC i.v. injections. TAC + antagomir and TAC + agomir were generated by i.v. injection of antagomir and agomir (30 pmol/g) 14 and 21 days before TAC surgery, respectively.
Click to Show/Hide
|
||||
Response regulation | MLK3 (MAP3K11) mainly regulates the JNK/ p53 signaling pathway-mediated oxidative stress and that ferroptosis causes myocardial fibrosis in the advanced stages of chronic heart failure (CHF). Promoting the expression of miR-351 can inhibit the expression of MLK3, and significantly improve cardiac function in mice subjected to TAC. | ||||
Experiment 28 Reporting the Ferroptosis Target of This Regulator | [46] | ||||
Responsed Disease | Health | ICD-11: N.A. | |||
Responsed Drug | Alumina | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hHCs (Hippocampal cells) | ||||
In Vivo Model |
Male healthy Wistar rats (six-week-old, provided by Experimental Animal Centre of Harbin Medical University, China) were used in this study. All rats (3-4 rats per cage) access to standard diet anddeionized waterad libitum and were placed in standard laboratory conditions. Seventy-two rats (weighing 200-220 g) were randomly divided into 4 groups (n = 18): AlNPs group was exposed to 50 mg/kg AlNPs (< 50nm, Sigma-Aldrich, USA) by gavage once a day for 90 days. CRS + AlNPs group was received CRS for 21 days and was exposed to 50 mg/kg AlNPs daily by gavage for 90 days. CRS + H2O group was subjected to CRS for 21 days and was given the same volume of deionized water daily by gavage for 90 days. The control (CON) group was given the same volume of deionized water daily and not affected by restraint stress for 90 days.
Click to Show/Hide
|
||||
Response regulation | Alumina nanoparticles (AlNPs) and CRS activated IFN-/ASK1/JNK (MAPK8) signaling pathway. Furthermore, IFN- neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. JNK inhibits ubiquitin-mediated p53 degradation by increasing phosphorylation of p53 at Ser6, which helps mediate oxidative stress to trigger ferroptosis. | ||||
Experiment 29 Reporting the Ferroptosis Target of This Regulator | [46] | ||||
Responsed Disease | Health | ICD-11: N.A. | |||
Responsed Drug | R46A2 | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hHCs (Hippocampal cells) | ||||
In Vivo Model |
Male healthy Wistar rats (six-week-old, provided by Experimental Animal Centre of Harbin Medical University, China) were used in this study. All rats (3-4 rats per cage) access to standard diet anddeionized waterad libitum and were placed in standard laboratory conditions. Seventy-two rats (weighing 200-220 g) were randomly divided into 4 groups (n = 18): AlNPs group was exposed to 50 mg/kg AlNPs (< 50nm, Sigma-Aldrich, USA) by gavage once a day for 90 days. CRS + AlNPs group was received CRS for 21 days and was exposed to 50 mg/kg AlNPs daily by gavage for 90 days. CRS + H2O group was subjected to CRS for 21 days and was given the same volume of deionized water daily by gavage for 90 days. The control (CON) group was given the same volume of deionized water daily and not affected by restraint stress for 90 days.
Click to Show/Hide
|
||||
Response regulation | Alumina nanoparticles (AlNPs) and CRS activated IFN-/ASK1/JNK (MAPK8) signaling pathway. Furthermore, IFN- neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. JNK inhibits ubiquitin-mediated p53 degradation by increasing phosphorylation of p53 at Ser6, which helps mediate oxidative stress to trigger ferroptosis. | ||||
Experiment 30 Reporting the Ferroptosis Target of This Regulator | [47] | ||||
Responsed Disease | Traumatic brain injury | ICD-11: NA07 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hBCs (Brain cells) | ||||
In Vivo Model |
The Experimental Animal Center at Southern Medical University provided the wild-type (WT) adult male and female C57BL/6 mice (body weight, 22-25 g). Briefly, mice were anesthetized with an intraperitoneal injection of sodium pentobarbital (30 mg/kg), and mounted on a stereotaxic frame. A midsagittal incision was made in the scalp and a circular craniotomy (4.5 mm diameter) was made over the left parietotemporal cortex.
Click to Show/Hide
|
||||
Response regulation | P53-mediated ferroptosis contributes to the pathogenesis of traumatic brain injury (TBI). Furthermore, SIRT2 exerts a neuroprotective effect against TBI by suppressing p53-mediated ferroptosis. | ||||
Transferrin receptor protein 1 (TFRC) [Driver; Suppressor; Marker]
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis Target of This Regulator | [28] | ||||
Target for Ferroptosis | Marker/Suppressor/Driver | ||||
Responsed Disease | Ischemia/reperfusion injury | ICD-11: DB98 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
Male Sprague-Dawley (SD) rats (250-300 g) were purchased from the Laboratory Animal Center, Xiangya School of Medicine, Central South University, China. Briefly, a left thoracotomy was carried out in the fourth intercostal space and the heart was exposed via opening the pericardium. The left coronary artery was around via a 4-0 silk suture and a snare was formed by passing both ends of the suture via a short polyethylene tubing. Blockage of the coronary artery was conducted via clamping the snare against the heart surface. Reperfusion was performed by release of the snare. The sham group conducted the same procedure but without ischemia (the snare was not tightened). To establish the I/R injury model, the rat hearts were subjected to 1 h-ischemia plus 3 h-reperfusion.
Click to Show/Hide
|
||||
Response regulation | A novel pathway of USP7/ p53/TfR1 has been identified in the ischemia/reperfusion (I/R)-treated rat hearts, where up-regulation of USP7 promotes ferrptosis via activation of the p53/TfR1 pathway. | ||||
Polyunsaturated fatty acid lipoxygenase ALOX15 (ALOX15) [Driver]
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis Target of This Regulator | [29] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Spinal cord injury | ICD-11: ND51 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
PC12 cells | Adrenal gland pheochromocytoma | Rattus norvegicus | CVCL_0481 | |
In Vivo Model |
Forty female Sprague-Dawley rats (200-300 g, 8 weeks old) were purchased from the Animal Experiment Center of Fudan University. Forty rats were randomly divided into four groups: sham operation group (n = 10), SCI group (n = 10), SCI + ferroptosis inhibitor group (SCI + ferrostatin1) (n = 10), and SCI + DHODH Inhibitor group (SCI + teriflunomide) (n = 10). Ten rats in the sham group only received laminectomy without SCI. To induce spinal cord injury, spinal cord injury surgery was performed in the middle thoracic region of rats (T8-T9).
Click to Show/Hide
|
||||
Response regulation | The application of DHODH is a potential treatment for spinal cord injury (SCI). DHODH can reduce the ferroptosis of neurons after spinal cord injury by regulating the P53/ALOX15 signaling pathway, thereby alleviating spinal cord injury. | ||||
Mitoferrin-2 (SLC25A28) [Driver]
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis Target of This Regulator | [33] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Liver fibrosis | ICD-11: DB93 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
hHSCs (Human hepatic stellate cells) | ||||
HSC-T6 cells | Normal | Rattus norvegicus | CVCL_0315 | ||
In Vivo Model |
Eight-week-old male C57BL/6 mice were purchased from Nanjing Medical University (Nanjing, China). Sixty mice were randomly divided into six groups of ten animals each with comparable mean body weight. Mice of six groups were treated with Sham, BDL + VA-Lip-Control-shRNA, BDL + VA-Lip-Control-shRNA + erastin, BDL + VA-Lip-BRD7-shRNA + erastin, BDL + VA-Lip-P53-shRNA + erastin or BDL + VA-Lip-SLC25A28-shRNA + erastin, respectively. Mice were anesthetized with isoflurane. A midline laparotomy was performed, and the common bile duct was ligated close to the liver hilus immediately below the bifurcation with 3-0 surgical silk and cut between the ligatures as described previously.
Click to Show/Hide
|
||||
Response regulation | BRD7- P53-SLC25A28 axis involves in mediating ferroptosis via mitochondrial iron metabolism pathway. These findings reveal novel signal transduction and regulatory mechanism of ferroptosis, and also suggest BRD7- P53-SLC25A28 axis as potential targets for liver fibrosis. | ||||
Glioblastoma [ICD-11: 2A00]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [4] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Pseudolaric acid B | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
U-87MG cells | Glioblastoma | Homo sapiens | CVCL_0022 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
SHG-44 cells | Astrocytoma | Homo sapiens | CVCL_6728 | ||
In Vivo Model |
Twenty athymic BALB/c nude mice (aged 4 weeks, weight 20-22 g, from Shanghai laboratory animal Center, Shanghai, China) were housed in a specific pathogen-free environment. A total of 1 x 106 logarithmically growing C6 cells in 100 uL of PBS were subcutaneously injected into the right flank of each mouse. Therapeutic experiments were started when the tumor reached about 150 mm3 after about 7 days. The mice were allocated to receive intraperitoneal injections of vehicle (control group, n = 5/group), PAB at the dosage of 10 mg/kg body weight (n = 10/group) and 20 mg/kg body weight (n = 10/group) in the same volume 50 uL once a days for 8 times.
Click to Show/Hide
|
||||
Response regulation | Pseudolaric acid B (PAB) improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT (SLC7A11) pathway, which further exacerbated accumulation of H2O2and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment. | ||||
T-cell lymphoma [ICD-11: 2B01]
In total 1 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [5] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Kayadiol | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
YT cells | Natural killer cell lymphoblastic leukemia | Homo sapiens | CVCL_1797 |
hPBLs (Human peripheral blood lymphocytes) | ||||
Response regulation | Kayadiol decreased the expression of SLC7A11 and GPX4, the negative regulatory proteins for ferroptosis. And p53 was the key mediator of kayadiol-induced ferroptosis by SLC7A11/GPX4 axis through p53 knockout experiments. Kayadiol can serve as an effective alternative in the treatment of NK/T cell lymphoma. | |||
Osteosarcoma [ICD-11: 2B51]
In total 2 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [6] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Bavachin | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
MG-63 cells | Osteosarcoma | Homo sapiens | CVCL_0426 |
HOS cells | Osteosarcoma | Homo sapiens | CVCL_0312 | |
Response regulation | Bavachin could induce Osteosarcoma cell ferroptosis. Furthermore, bavachin elevated intracellular ferrous iron levels by increasing TFRC and DMT1 expression and decreasing FTH and FTL expressions. Bavachin also reduced SLC7A11 and GPX4 expression and promoted ROS and MDA accumulation by downregulating p-STAT3 to upregulate P53 expression. | |||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [36] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model |
U2OS cells | Osteosarcoma | Homo sapiens | CVCL_0042 |
IMR-90 cells | Normal | Homo sapiens | CVCL_0347 | |
Response regulation | SOCS1 impacts the pattern of secreted products in cells with active p53 and is required for the expression of a selective set of p53 target genes including those involved in ferroptosis. SOCS1 can use several mechanisms to activate p53, including promotion of serine-15 phosphorylation by ATM/ATR kinases and inhibition of the p53 repressor KAP1 in osteosarcoma cell lines. | |||
Oral squamous cell carcinoma [ICD-11: 2B6E]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [1] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Quisinostat | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Necroptosis | hsa04217 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell apoptosis | |||||
Cell pyroptosis | |||||
In Vitro Model |
CAL-27 cells | Tongue adenosquamous carcinom | Homo sapiens | CVCL_1107 | |
Tca8113 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6851 | ||
In Vivo Model |
Adult male athymic BALB/c nude mice (20-22 g of 5-week-old mice) were housed in a controlled environment at 23 ± 2 and 40%-70% humidity under a 12 h dark/light cycle with free access to irradiated food and sterile water. A suspension of 6 x 106/100 uL TCA-8113 cells was inoculated subcutaneously into the hind flank region of each nude mouse. The average tumor volume in nude mice reached 100 mm3, and mice were randomly divided into three groups. Quisinostat was formulated in normal saline and administered at 3 and 10 mg/kg/day byintraperitoneal injection. Control mice were given equal volume saline intraperitoneally. The tumor volume and the bodyweight of mice were monitored every three days.
Click to Show/Hide
|
||||
Response regulation | Quisinostat could increase the apoptosis rate in the tumor tissues of nude mice. Up-regulation of the expression of p53 and down-regulated expression of GPX4 in cell lines were observed by immunofluorescent staining, and the expression locations of p53 and GPX4 proteins in TSCC cells were observed. Quisinostat may be a potential drug for the treatment of tongue squamous cell carcinoma. | ||||
Gastric cancer [ICD-11: 2B72]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [7] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Tanshinone IIA | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
BGC-823 cells | Gastric carcinoma | Homo sapiens | CVCL_3360 | |
NCI-N87 cells | Gastric tubular adenocarcinoma | Homo sapiens | CVCL_1603 | ||
In Vivo Model |
All mice were housed under a setting of 12-h light/dark cycle at 22 ± 1, 55% humidity and fed with water and food provided at regular time. During the entire maintenance period, all mice were permitted free cage activity without joint immobilization. The initial body weights of the mice were between 20 and 23 grams. After subcutaneous injection of 2 x 106 BGC-823 gastric cancer cells into the back of NOD-SCID mice, the mice were treated with or without Tan IIA (50 mg/kg) or Tan IIA in combination with Fer-1 (50 mg/kg). Tan IIA was diluted in DMSO:Methanol:Hydroxypropyl-b-cydodextrin (HP-b-CD) = 1:1:1. Fer-1 was also dissolved in DMSO:Methanol:HP-b-CD. Seven days after BGC-823 gastric cancer cells injection, intraperitoneal injection with Tan IIA was carried out every other day followed by killing at day 22 of tumor cell inoculation. All mice were killed by dislocation of the cervical vertebrae. Before killing, the tumor volume was measured every 3 days. All experiments were carried out using six mice each group in three independent experiments of a time-dependent manner with three time points.
Click to Show/Hide
|
||||
Response regulation | Tanshinone IIA increased lipid peroxidation and up-regulated Ptgs2 and Chac1 expression, two markers of ferroptosis. In addition, Tan IIA also up-regulated p53 expression and down-regulated xCT (SLC7A11) expression. Therefore, Tan IIA could suppress the proliferation of gastric cancer via inducing p53 upregulation-mediated ferroptosis. | ||||
Melanoma [ICD-11: 2C30]
In total 1 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [8] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Gambogenic Acid | Investigative | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell adhesion molecules | hsa04514 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
Cell migration | ||||
Cell invasion | ||||
In Vitro Model |
A-375 cells | Amelanotic melanoma | Homo sapiens | CVCL_0132 |
A2058 cells | Amelanotic melanoma | Homo sapiens | CVCL_1059 | |
Response regulation | Gambogenic acid (GNA) significantly inhibited the invasion, migration and EMT in melanoma cells, and these cells exhibited small mitochondrial wrinkling (an important feature of ferroptosis). GNA upregulated the expression of p53, solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) in the model cells, contributing to the mechanisms underlying GNA-induced ferroptosis. | |||
Parkinson disease [ICD-11: 8A00]
In total 3 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [9] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Cyperquat | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
PC12 cells | Adrenal gland pheochromocytoma | Rattus norvegicus | CVCL_0481 | |
Response regulation | As a classic drug employed inin vitromodels of Parkinson's disease, 1-methyl-4-phenylpyridinium (MPP) can induce senescence in PC12 cells. The expression of the ferroptosis-related proteins ASCL4 was upregulated and FTH1 was downregulated, which promoted accumulation of lipid peroxides and eventually led to ferroptosis. By rescuing MPP-induced ferroptosis, cell senescence could be inhibited, and its molecular mechanism was related to a p53/SLC7A11/GPX4 signaling pathway. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [23] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Clioquinol | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
PI3K-Akt signaling pathway | hsa04151 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
SK-N-SH cells | Neuroblastoma | Homo sapiens | CVCL_0531 | |
In Vivo Model |
In total, twelve healthy adult rhesus monkeys (Macaca mulatta lasiotis, aged 4-5 years, and weighed 3.5-5 kg at the start of the study) were obtained from Sichuan Primed Biological Technology Co., Ltd. Monkeys were randomly divided into two groups: normal (control) group (n = 3) and MPTP group (n = 9). Monkeys from MPTP group were administered with MPTP by intramuscular injection daily at the beginning of the study, and then the MPTP dose was gradually added to 0.5 mg/kg at the end of the experiment. Monkeys from control group were injected with saline instead, and the other conditions were the same with MPTP group.
Click to Show/Hide
|
||||
Response regulation | Ferroptosis was probably involved in the pathogenesis of parkinson's disease (PD). Clioquinol (CQ) can decrease the excessive iron in the SN to normal level and directly protect DA neurons against oxidative stress probably by activating the AKT/mTOR survival pathway and blocking p53-medicated cell death. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response | [24] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Ferric ammonium citrate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
MES23.5 cells | Normal | Mus musculus | CVCL_J351 | |
In Vivo Model |
Human a-synuclein (a-Syn) A53T overexpressiontransgenic mice(B6; C3-Tg (Prnp-SNCA*A53T) 83Vle/J) were originally obtained in breeding pairs from the Jackson Laboratory (004479) to generate a stable breeding colony. Animals were raised according to SPF level, kept at constant temperature (20 ± 2) , constant humidity (50 ± 10%), day and night cycle light (12-12 h), with free access to food and water.
Click to Show/Hide
|
||||
Response regulation | Ferroptosis firstly occurred in a relatively low concentration of ferric ammonium citrate (FAC)-treated group, and then apoptosis appeared in response to the increased iron doses. This was also confirmed in vivo in parkinson's disease transgenic mice and the underlying mechanism might be associated with the p53 signaling pathway, but not MAPK signaling pathway. | ||||
Status epilepticus [ICD-11: 8A66]
In total 2 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [2] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Apigenin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
SH-SY5Y cells | Neuroblastoma | Homo sapiens | CVCL_0019 | |
In Vivo Model |
5-weeks-old kainate (KA)-induced BALB/c nude mice, a widely used epilepsy mouse model, were performed with intraperitoneal (i.p.) injection of KA (6 mg/kg). Pre-treatment 21 with antioxidant apigenin (60 mg/Kg, 2 days) or post-treatment with apigenin (60 mg/Kg, 1 day), mice were injected with KA (6 mg/kg) via intraperitoneal (i.p.) injection, and then HCP (0.5 mg/Kg) were injected by intravenous (i.v.) injection. In vivo and Ex vivo fluorescence images of relative ClO levels in mice brains 5, 15, 30, 45, and 60 min post injection of HCP were further performed by using the IVIS Spectrum imaging system (Nanjing University) with an excitation filter of 430 nm and the collection wavelength range is from 500-600 nm.
Click to Show/Hide
|
||||
Response regulation | Apigenin can efficiently reduce the expression of intracellular MPO and increase the levels of GPX4 and SIRT1, thereby conferring neuroprotection through regulation of kainic acid (KA)-induced ferroptosis. And the level of Ac-p53 inside the brains treated with apigenin was down-regulated, suggesting that the p53-mediated ferroptosis pathway might be blocked. Overall, apigenin was screened and confirmed as an efficient lead compound for epilepsy prevention and treatment. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [10] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Seratrodast | Discontinued in Phase 3 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
HT22 cells | Normal | Mus musculus | CVCL_0321 | |
In Vivo Model |
Drugs were dissolved in vehicle (0.1% DMSO + 20% PEG 300 + 0.5% CMC-Na + ddH2O). Mice in Control and PTZ groups were administered for five days with an equivalent volume of vehicle. PTZ-induced seizure model was done for the subsequent 1 h after the last administration of drugs. We performed a preliminary doseresponse trial, the dose of 60 mg/kg was established as being sufficient to trigger seizures with lower mortality and chosen as the optimal dose. One mouse in PTZ group was dead due to a severe seizure. At the end of the experiment, the mice were anesthetized or euthanized. For histopathological studies, the mice were anesthetized and intracardially perfused with 0.9% saline, followed by 0.4% paraformaldehyde for fixation of the brain. For immunoblot analysis, the hippocampus was rapidly isolated.
Click to Show/Hide
|
||||
Response regulation | Seratrodast could reduce lipid ROS production, regulate the system xc-/glutathione (GSH)/glutathione peroxidase 4 (GPX4) axis, and inhibit JNK (MAPK8) phosphorylation and p53 expression. JNK can directly or indirectly modulate the expression and activation of p53, which could regulate ferroptosis through inhibition of SLC7A11 transcription. Seratrodast increased the latency of seizures and reduced seizure duration in pentylenetetrazole-induced seizures in Epilepsy. | ||||
Intracerebral hemorrhage [ICD-11: 8B00]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [11] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Isorhynchophylline | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
p53 signaling pathway | hsa04115 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
HT22 cells | Normal | Mus musculus | CVCL_0321 | |
In Vivo Model |
Adult male Sprague-Dawley rats (SD rats, weighing 250-300 g) aged 11-12 weeks were purchased from SLAC Laboratory Animal Co., Ltd. (Shanghai, China). All 96 rats were randomly divided into four groups of 24 rats each: Sham group, Sham + IRN (30 mg/Kg) group, ICH group, and ICH + IRN (30 mg/Kg) group. The rats in sham group were injected with PBS solution, and the Sham + IRN (30 mg/Kg) group was received an equal amount of 30 mg/Kg IRN solution (intra-peritoneal injection) after the sham operation. After ICH, the rats in ICH group were injected with PBS solution, and the ICH + IRN (30 mg/Kg) group was received an equal amount of 30 mg/Kg IRN solution (intra-peritoneal injection).
Click to Show/Hide
|
||||
Response regulation | Isorhynchophylline (IRN) decreased ferroptosis and lipid ROS level, upregulated the expression of miR-122-5p and SLC7A11 mRNA, and inhibited TP53 expression. In conclusion, IRN protects neurocyte from intracerebral hemorrhage (ICH)-induced ferroptosis via miR-122-5p/TP53/SLC7A11 pathway, which may provide a potential therapeutic mechanism for ICH. | ||||
Sarcopenia [ICD-11: FB32]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [12] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Ferric citrate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Glutathione metabolism | hsa00480 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
C2C12 cells | Normal | Mus musculus | CVCL_0188 | |
In Vivo Model |
The 8-week- and 40-week-old male SAMP8 mice were purchased from the model animal research center of Zhishan Institute of Healthcare Research Co., Ltd. (Beijing, China). All the mice were kept in an SPF grade animal facility at 24 with a relative humidity of 50%-60%, and in a light/dark cycle of 12 h/12 h.
Click to Show/Hide
|
||||
Response regulation | Ferric citrate induced ferroptosis in C2C12 cells, as well as impaired their differentiation from myoblasts to myotubes. Iron overload upregulated the expression of P53, which subsequently repressed the protein level of Slc7a11 (solute carrier family 7, member 11), a known ferroptosis-related gene. Targeting iron accumulation and ferroptosis might be a therapeutic strategy for treating sarcopenia. | ||||
Neurotoxicity [ICD-11: NE61]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [3] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Sevoflurane | Approved | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hBCs (Brain cells) | ||||
In Vivo Model |
Pregnant rats were placed in a dedicated plastic chamber with ambient gas at a flow rate of 2L/min. Fer-1 solubilized in saline and 1% dimethyl sulfoxide (DMSO) and PD146176 (a specific 15LOX inhibitor) dissolved in corn oil containing 1% DMSO were administered intraperitoneally to rats at a dose of 5 mg/kg 1 h before each exposure, respectively. Similarly, 0.5 mg/kg Ku55933 (an ATM inhibitor), which is diluted in saline containing 1% DMSO, was intraperitoneally administered 2 h previously.
Click to Show/Hide
|
||||
Response regulation | Sevoflurane could enhance 15LO2-PEBP1 interaction and activate ATM and its downstream P53/SAT1 pathway, which might be attributed to excessive p-ATM nuclear translocation, indicating a potential therapeutic target for ameliorating sevoflurane-induced neurotoxicity. | ||||
HIV Infection [ICD-11: 1C60]
In total 1 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [13] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Methamphetamine | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
BV-2 cells | Normal | Mus musculus | CVCL_0182 |
Response regulation | Methamphetamine (METH) and HIV-1 lead to oxidative stress and their combined effect increases the risk of HIV-associated neurocognitive disorder (HAND), which may be related to the synergistic ferroptotic impairment in microglia. We found that METH and HIV-1 Tat reduced the expression of ferroptotic protein GPX4 and the cell viability and enhanced the expression of P53 and the level of ferrous iron, while the above indices were significantly improved with pretreatment of ferrostatin-1. | |||
Colon cancer [ICD-11: 2B90]
In total 5 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [14] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Albiziabioside A | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
MCF-7 cells | Breast carcinoma | Homo sapiens | CVCL_0031 | |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | ||
hBMECs (Human brain microvascular endothelial cells) | |||||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
HaCaT cells | Normal | Homo sapiens | CVCL_0038 | ||
HCT-8 cells | Ileocecal adenocarcinoma | Homo sapiens | CVCL_2478 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
CCD-841CoN cells | Normal | Homo sapiens | CVCL_2871 | ||
BEAS-2B cells | Normal | Homo sapiens | CVCL_0168 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
In Vivo Model |
A total of 1 x 107 HCT116 cells were subcutaneously inoculated into the right flank of BALB/c mice. When tumor reached 70-100 mm3 (10 days after implant), mice were divided into five groups of eight animals at random. The groups with D13 were administered intravenously 20 mg/kg and 10 mg/kg. The positive control group was treated with AlbA(20 mg/kg and 10 mg/kg) through intravenous injection. The negative control group received 0.9% normal saline through intravenous injection.
Click to Show/Hide
|
||||
Response regulation | The study synthesized a series of Albiziabioside A derivatives and evaluated the antitumor activity both in vitro and in vivo. Compound D13 could induce apoptosis and ferroptosis through the mitochondrial pathway as a p53 activator. In addition, compound D13 significantly suppressed tumorigenesis without inducing toxicity in normal organs in vivo. The antitumor efficacy of D13 was further verified in colon cancer xenograft mouse models. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [15] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Ginkgo biflavones | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
HT29 cells | Colon cancer | Mus musculus | CVCL_A8EZ | ||
RKO cells | Colon carcinoma | Homo sapiens | CVCL_0504 | ||
LoVo cells | Colon adenocarcinoma | Homo sapiens | CVCL_0399 | ||
SW620 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0547 | ||
In Vivo Model |
HCT-116 cells (5 x 106) were injected into the flanks of 6-week-old male BALB/c nude mice to generate xenografts. The mice were randomly divided into four groups (n = 7 per group), and treatment was started at 96 h postinjection. The mice received an intraperitoneal injection (i.p.) of 0.9% saline solution containing 5% dimethyl sulfoxide (DMSO) (vehicle for blank control) and ginkgetin (10 mg/kg) once a day, respectively, and 5-FU (30 mg/kg) was used as the positive control (i.p., once every 3 days, alternately).
Click to Show/Hide
|
||||
Response regulation | Ginkgo biflavones can increase the expression level of p53 by inhibiting the expression of MDM2 protein and induce cell death independent of p53 transcriptional activity in vitro. And we provide evidence that ginkgetin strengthened the antitumor effect of fluorouracil (5-FU) in the HCT-116 colon cancer xenograft model. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response | [30] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
HeLa cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0030 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
HIEC-6 cells | Normal | Homo sapiens | CVCL_6C21 | ||
Response regulation | RRM1 increases the instability of p53 by regulating the physical interaction of p53 with the ubiquitinating enzyme MDM2 and the deubiquitinating enzyme USP11, subsequently suppressing p21 (CDKN1A) and GPX4 in colon carcinoma cells, thereby promoting the accumulation of lipid peroxidation and occurrence of radiation-induced ferroptosis. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response | [37] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CT26 cells | Colon adenocarcinoma | Mus musculus | CVCL_7254 | |
4T1 cells | Mammary carcinoma | Mus musculus | CVCL_0125 | ||
In Vivo Model |
BALB/c and C57BL/6 mice were obtained from the Jackson Laboratory (Bar Harbor, ME). To establish subcutaneous tumor models, CT26 cells (2 x 105 cells/mouse) were injected into the right flanks of BALB/c mice. For experimental lung metastasis models, colon carcinoma CT26, mesothelioma AB1 (2 x 105 cells/mouse) and mammary carcinoma 4T1 (2 x 104 cells/mouse) were injected into BALB/c mice. Tumor-bearing mice were treated with vehicle PEG300 (Sigma-Aldrich) and NC06 (dissolved in PEG300), respectively, every two days for 3-5 times by Intraperitoneal injection.
Click to Show/Hide
|
||||
Response regulation | ASAH2 is overexpressed in MDSCs in human colon cancer patients. ASAH2 protects MDSCs from ferroptosis through destabilizing p53 protein to suppress the p53 pathway in MDSCs in the tumor microenvironment. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response | [38] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Hippo signaling pathway | hsa04390 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
SW620 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0547 | ||
Response regulation | CYGB significantly increased the sensitivity of cancer cells to RSL3- and erastin-induced ferroptotic cell death. Collectively, a novel tumour suppressor role of CYGB through p53-YAP1 axis in regulating ferroptosis and suggested a potential therapeutic approach for colon cancer. | ||||
Colorectal cancer [ICD-11: 2B91]
In total 2 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [16] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | BEBT-908 | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
PI3K-Akt signaling pathway | hsa04151 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
MC-38 cells | Colon adenocarcinoma | Homo sapiens | CVCL_B288 | |
In Vivo Model |
About 2 x 105 MC38 tumor cells were inoculated subcutaneously into C57BL/6 mice. A total of 100 mg/kg BEBT-908 was intravenously injected every other day for four times initiating from day 4 after tumor cell inoculation. Tumors were harvested on day 12 after inoculation, weighted, mechanically minced and incubated with 50 ug/mL DNase I (Sigma) and 2 mg/mL collagenase P (Sigma) for 20 minutes at 37.
Click to Show/Hide
|
||||
Response regulation | Treatment with BEBT-908 promoted ferroptotic cell death of cancer cells by hyperacetylating p53 and facilitating the expression of ferroptotic signaling. The dual PI3K/HDAC inhibitor BEBT-908 elicits potent antitumor responses, effectively inducing immunogenic ferroptosis of colorectal cancer cells and potentiating cancer immunotherapy. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [31] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Ferroptosis | hsa04216 | |||
MAPK signaling pathway | hsa04010 | ||||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
SW480 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0546 | ||
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | ||
In Vivo Model |
Six 4-week-old male BALB/c nude mice were ordered from the Shanghai Laboratory Animal Center (Shanghai SLAC Laboratory Animal Co., Ltd., China). A total of 5 x 106 TIPE+/+ SW480 cells were suspended in 100 uL of PBS and subcutaneously injected into the right axilla flank of each nude mouse, and the same amount of vector SW480 cells was into the left. At 2 weeks after inoculation, the xenograft tumor size was measured using Vernier calipers every 2 days.
Click to Show/Hide
|
||||
Response regulation | MiR-539 can bind to and regulate the expression of TIPE, and miR-539 activates SAPK/JNK to downregulate the expression of glutathione peroxidase 4 (GPX4) and promote ferroptosis. In addition, SAPK/JNK is the upstream molecule of p53. MiR-539 is a new therapeutic target for colorectal cancer (CRC) patients. | ||||
Pancreatic cancer [ICD-11: 2C10]
In total 2 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [17] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
Panc02 cells | Pancreatic ductal adenocarcinoma | Mus musculus | CVCL_D627 | |
PANC-1 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0480 | ||
In Vivo Model |
Six to eight-week-old female C57BL/6 mice were purchased from the Experimental Animal Center of Military Medical Sciences (Beijing, China). C57BL/6 mice were anesthetized and the tail of the pancreas was exposed. Panc 02 cells were resuspended in PBS at a concentration of 1 x 106 cells/0.1 ml and 50 ul cells were injected into the tail of the pancreas. Tumor-bearing mice were randomly divided into two groups (3 days after implantation). The control group was intraperitoneally injected 200 ul PBS daily for 10 days, and the DHA group was intraperitoneally injected with 100 mg/kg DHA daily for 10 days. The pancreatic tumors and spleens of the mice were collected for subsequent analysis.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin has anti-tumor effect in pancreatic cancer cells in vitro and in vivo. DHA treatment induced ferroptosis by increasing P53 and AOLX12 expression. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [32] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
BxPC-3 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0186 | |
PANC-1 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0480 | ||
Response regulation | Cold induction promotes the process of ferroptosis by inducing the expression of CIRBP and then regulating key factors such as p53 and GPX4[. In addition, cold induction significantly inhibited the proliferation of pancreatic cancer cells and induced cell apoptosis, but after the addition of ferroptosis inhibitor, cell proliferation and apoptosis did not change significantly. | ||||
Lung cancer [ICD-11: 2C25]
In total 3 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [18] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Levobupivacaine | Approved | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model |
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | |
A427 cells | Lung carcinoma | Homo sapiens | CVCL_1055 | ||
In Vivo Model |
Balb/c nude mice (n= 5, 4-week-old, male) were applied to detect the impact of levobupivacaine on tumor growth. Mice subcutaneously injected with 1 x 107 A549 cells were treated with levobupivacaine (40 umol/Kg) or equal volume saline. The tumor volume was remarked every 5 days and finished at 30 days after injection, followed by the analysis of volume (length (width/2)2) and weight.
Click to Show/Hide
|
||||
Response regulation | Levobupivacaine could inhibit the proliferation and induce the apoptosis of non-small cell lung cancer (NSCLC) cells. Levobupivacaine was able to attenuate the invasion and migration in the cells. The treatment of levobupivacaine remarkably increased the levels of ROS, iron, and Fe2+ in NSCLC cells. Mechanically, levobupivacaine up-regulated the expression of p53 and induced ferroptosis by regulating p53 in NSCLC cells. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [40] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Cell adhesion molecules | hsa04514 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
NCI-H460 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0459 | ||
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
NCI-H358 cells | Minimally invasive lung adenocarcinoma | Homo sapiens | CVCL_1559 | ||
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
In Vivo Model |
The 4-wk-old female nude mice used in this study were purchased from Hunan SJA Laboratory Animal Co., Ltd (Changsha). Then, 2 x 106 GINS4 WT or KO A549 cells were injected s.c. into nude mice without matrigel. Tumor size was measured every 2 d with a caliper, and volume of tumor was calculated with the formula: L x W2 x 0.5, for L represents the longest diameter and W means the shortest diameter. Then, 2 x 106 p53 WT and KO A549 cells with shCon or shGINS4 overexpressing were injected s.c. into nude mice without matrigel. When tumors reached 60 to 100 mm3, p53 WT with shGINS4 mice were treated with ferrostatin-1 (S7243, Selleckchem) (20 mg/kg) in 2% DMSO, 50% PEG300, 5% Tween80, and 43% water by daily intraperitoneal injection. Tumors were measured three times a week. Mice were sacrificed and tumors were collected finally. Tumor tissue was made into a single-cell suspension for lipid ROS assay using Tumor Dissociation Kit (Miltenyi Biotec).
Click to Show/Hide
|
||||
Response regulation | GINS4 is a potential oncogene in lung adenocarcinoma (LUAD) that functions to destabilize p53 and then inhibits ferroptosis, providing a potential therapeutic target for LUAD. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response | [41] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | |
SPC-A1 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6955 | ||
NCI-H522 cells | Non-small cell lung carcinoma | Homo sapiens | CVCL_1567 | ||
HBE1 cells | Normal | Homo sapiens | CVCL_0287 | ||
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
MRC-5 cells | Normal | Homo sapiens | CVCL_0440 | ||
NCI-H358 cells | Minimally invasive lung adenocarcinoma | Homo sapiens | CVCL_1559 | ||
PC-9 cells | Lung adenocarcinoma | Homo sapiens | CVCL_B260 | ||
95C cells | Lung giant cell carcinoma | Homo sapiens | CVCL_7109 | ||
95D cells | Lung giant cell carcinoma | Homo sapiens | CVCL_7110 | ||
In Vivo Model |
SCID mice (Hunan SJA Laboratory Animal Co. Ltd.) were injected with the indicated cells in the mammary fat pad (10 mice/group). Injected mice were imaged from both the dorsal and ventral sides every three days.
Click to Show/Hide
|
||||
Response regulation | P53RRA bound Ras GTPase-activating protein-binding protein 1 (G3BP1) using nucleotides 1 and 871 of P53RRA and the RRM interaction domain of G3BP1 (aa 177-466). The cytosolic P53RRA-G3BP1 interaction displaced p53 from a G3BP1 complex, resulting in greater p53 retention in the nucleus, which led to cell-cycle arrest, apoptosis, and ferroptosis in lung cancer cell lines. | ||||
Ovarian cancer [ICD-11: 2C73]
In total 2 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [19] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Apatinib | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
Cell autophagy | ||||
In Vitro Model |
A2780 cells | Ovarian endometrioid adenocarcinoma | Homo sapiens | CVCL_0134 |
OVCAR-3 cells | Ovarian serous adenocarcinoma | Homo sapiens | CVCL_0465 | |
Response regulation | Apatinib combined with olaparib-induced ferroptosis via a p53-dependent manner in ovarian cancer. Further studies showed that apatinib combined with olaparib-induced ferroptosis by inhibiting the expression of Nrf2 and autophagy, thereby inhibiting the expression of GPX4. The Nrf2 activator RTA408 and the autophagy activator rapamycin rescued the combination drug-induced ferroptosis. | |||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [19] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Olaparib | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
Cell autophagy | ||||
In Vitro Model |
A2780 cells | Ovarian endometrioid adenocarcinoma | Homo sapiens | CVCL_0134 |
OVCAR-3 cells | Ovarian serous adenocarcinoma | Homo sapiens | CVCL_0465 | |
Response regulation | Apatinib combined with olaparib-induced ferroptosis via a p53-dependent manner in ovarian cancer. Further studies showed that apatinib combined with olaparib-induced ferroptosis by inhibiting the expression of Nrf2 and autophagy, thereby inhibiting the expression of GPX4. The Nrf2 activator RTA408 and the autophagy activator rapamycin rescued the combination drug-induced ferroptosis. | |||
Prostate cancer [ICD-11: 2C82]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [20] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Flubendazole | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell apoptosis | |||||
In Vitro Model |
PC-3 cells | Prostate carcinoma | Homo sapiens | CVCL_0035 | |
DU145 cells | Prostate carcinoma | Homo sapiens | CVCL_0105 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
RWPE-1 cells | Normal | Homo sapiens | CVCL_3791 | ||
In Vivo Model |
24 nude mice (3-4 weeks) were acquired from the experimental animal center of southern medical university (GuangZhou, China) and kept under specific pathogen-free conditions. 4 x 106 PC3 cells were implanted subcutaneously into the right armpit regions of each nude mouse. When the tumors volume reached approximately 40 mm3, mice were randomly divided into two groups to receive flubendazole (10 mg/kg, once daily), 5-fluorouracil (30 mg/kg, once daily), their combination, and vehicle control by intraperitoneal injection. After 20 days of treatment, all mice were sacrificed and tumor weight and tumor volume were immediately measured, respectively.
Click to Show/Hide
|
||||
Response regulation | Flubendazole is a novel P53 inducer which exerts anti-proliferation and pro-apoptosis effects in castration-resistant prostate cancer (CRPC) through hindering the cell cycle and activating the ferroptosis, and indicates that a novel utilization of flubendazole in neoadjuvant chemotherapy of CRPC. | ||||
Hereditary Leiomyomatosis [ICD-11: 2C90]
In total 1 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [21] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Artesunate | Investigative | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model |
Caki-1 cells | Clear cell renal cell carcinoma | Homo sapiens | CVCL_0234 |
786-O cells | Renal cell carcinoma | Homo sapiens | CVCL_1051 | |
KTCTL-26 cells | Clear cell renal cell carcinoma | Homo sapiens | CVCL_5872 | |
A-498 cells | Renal cell carcinoma | Homo sapiens | CVCL_1056 | |
Response regulation | Artesunate (ART) significantly increased cytotoxicity and inhibited proliferation and clonogenic growth in both parental and sunitinib-resistant renal cell carcinoma (RCC) cells. P53 exclusively appeared in the KTCTL-26 cells, indicating that p53 might be predictive for ART-dependent ferroptosis. Thus, ART may hold promise for treating selected patients with advanced and even therapy-resistant RCC. | |||
Adrenocortical carcinoma [ICD-11: 2D11]
In total 1 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [22] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Romidepsin | Investigative | ||
Pathway Response | Cell adhesion molecules | hsa04514 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model |
SW13 cells | Adrenal cortex carcinoma | Homo sapiens | CVCL_0542 |
Response regulation | HDAC inhibitor Romidepsin converted SW13+ cells also had reduced mRNA expression of the mitochondrial ROS detoxifier superoxide dismutase 2 (SOD2), and the tumor suppressor p53. HDAC inhibitor treatment synergistically increased adrenocortical carcinoma cell death following induction of ferroptosis. | |||
Cardiomyopathy [ICD-11: BC43]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [25] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Berberine | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
All animal experiment protocols were implemented in accordance with the National Institutes of Health (NIH) guidelines, and the procedures were approved by the Animal Ethics Committee of Southwest University. C57BL/6J male mice, 8-10 weeks old, weighing 20 ± 2 g, were used in this study. Mice were housed under standard conditions at 22-24 with a 12 h light/12 h darkness cycle and free access to food and tap water. Thirty-six mice were randomly divided into six groups: control (N = 8), IMA group (50 mg/kg) (N = 8), Low-Ber (20 mg/kg) + IMA group (N = 8), Medium-Ber (40 mg kg1) + IMA group (N = 8), High-Ber (80 mg/kg) + IMA group (N = 8), and Fer-1 (1 mg/kg) + IMA group (N = 8). IMA was given intraperitoneally for 14 days. Ber was given orally 2 h before IMA treatment and Fer-1 was given intraperitoneally 2 h before IMA treatment.
Click to Show/Hide
|
||||
Response regulation | Berberine downregulated the expression of transferrin receptor (TfR) and P53 and upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), ferritin heavy chain-1 (FTH1), and glutathione peroxidase 4 (GPX4) in H9c2 cells and mice. The present data indicated that Ber has the potential to protect against IMA-induced cardiotoxicity, partlyviainhibiting Nrf2-dependent ferroptosis. | ||||
Liver fibrosis [ICD-11: DB93]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [33] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model |
hHSCs (Human hepatic stellate cells) | ||||
HSC-T6 cells | Normal | Rattus norvegicus | CVCL_0315 | ||
In Vivo Model |
Eight-week-old male C57BL/6 mice were purchased from Nanjing Medical University (Nanjing, China). Sixty mice were randomly divided into six groups of ten animals each with comparable mean body weight. Mice of six groups were treated with Sham, BDL + VA-Lip-Control-shRNA, BDL + VA-Lip-Control-shRNA + erastin, BDL + VA-Lip-BRD7-shRNA + erastin, BDL + VA-Lip-P53-shRNA + erastin or BDL + VA-Lip-SLC25A28-shRNA + erastin, respectively. Mice were anesthetized with isoflurane. A midline laparotomy was performed, and the common bile duct was ligated close to the liver hilus immediately below the bifurcation with 3-0 surgical silk and cut between the ligatures as described previously.
Click to Show/Hide
|
||||
Response regulation | BRD7- P53-SLC25A28 axis involves in mediating ferroptosis via mitochondrial iron metabolism pathway. These findings reveal novel signal transduction and regulatory mechanism of ferroptosis, and also suggest BRD7- P53-SLC25A28 axis as potential targets for liver fibrosis. | ||||
Ischemia/reperfusion injury [ICD-11: DB98]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [28] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
Male Sprague-Dawley (SD) rats (250-300 g) were purchased from the Laboratory Animal Center, Xiangya School of Medicine, Central South University, China. Briefly, a left thoracotomy was carried out in the fourth intercostal space and the heart was exposed via opening the pericardium. The left coronary artery was around via a 4-0 silk suture and a snare was formed by passing both ends of the suture via a short polyethylene tubing. Blockage of the coronary artery was conducted via clamping the snare against the heart surface. Reperfusion was performed by release of the snare. The sham group conducted the same procedure but without ischemia (the snare was not tightened). To establish the I/R injury model, the rat hearts were subjected to 1 h-ischemia plus 3 h-reperfusion.
Click to Show/Hide
|
||||
Response regulation | A novel pathway of USP7/ p53/TfR1 has been identified in the ischemia/reperfusion (I/R)-treated rat hearts, where up-regulation of USP7 promotes ferrptosis via activation of the p53/TfR1 pathway. | ||||
Osteonecrosis [ICD-11: FB81]
In total 1 item(s) under this disease | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [26] | |||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | ||
Responsed Drug | Zoledronic acid | Investigative | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
RAW 264.7 cells | Leukemia | Mus musculus | CVCL_0493 |
Response regulation | FBXO9 was downregulated in Zoledronic acid-treated osteoclast and promoted osteoclasts ferroptosis by inhibiting FBXO9-mediated p53 ubiquitination and degradation. The study provided a possible theoretical target for the clinical treatment of Bisphosphonates (BPs)-related osteonecrosis of jaw (BRONJ). | |||
Lung injury [ICD-11: NB32]
In total 2 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [27] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Lipopolysaccharide | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
MLE-12 cells | Normal | Mus musculus | CVCL_3751 | |
Response regulation | Silence of MLK3 (MAP3K11) alleviated Lipopolysaccharide (LPS)-induced lung epithelial cell injury by inhibiting p53-mediated ferroptosis, suggesting that MLK3 may be a potential target to prevent acute lung injury. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [34] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
THP-1 cells | Childhood acute monocytic leukemia | Homo sapiens | CVCL_0006 | |
HBE1 cells | Normal | Homo sapiens | CVCL_0287 | ||
In Vivo Model |
For the models of CS and LPS exposure, mice were anesthetized and intratracheally instilled with CS suspensions (3 mg/50 ul) or LPS (1 mg/kg). For the models of CS + Ferr-1/DFO, mice were intraperitoneally injected with Ferr-1 (1.25 umol/kg) or intranasal instilled with DFO (10 mg/kg) for 7 consecutive days after CS instillation. For the models of LPS + Ferr-1/DFO, mice were pretreated with Ferr-1 or DFO for 2 consecutive days and then intratracheally instilled with LPS. Mice were sacrificed 24 h after LPS instillation. For the X-ray exposure model, mice were exposed to ionizing radiation (IR) at 20 Gy, which was delivered at the dose rate of 2 Gy/min and a source skin distance of 51 cm by an X-ray generator (Model X-RAD320iX; Precision X-Ray, Inc., North Branford, CT, USA), and sacrificed 3 days after radiation.
Click to Show/Hide
|
||||
Response regulation | STAT6 negatively regulates ferroptosis through competitively binding with CBP, which inhibits P53 acetylation and transcriptionally restores SLC7A11 expression. Finally, pulmonary-specific STAT6 overexpression decreased the ferroptosis and attenuated CS and LPS induced acute lung injury. | ||||
Spinal cord injury [ICD-11: ND51]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [29] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
PC12 cells | Adrenal gland pheochromocytoma | Rattus norvegicus | CVCL_0481 | |
In Vivo Model |
Forty female Sprague-Dawley rats (200-300 g, 8 weeks old) were purchased from the Animal Experiment Center of Fudan University. Forty rats were randomly divided into four groups: sham operation group (n = 10), SCI group (n = 10), SCI + ferroptosis inhibitor group (SCI + ferrostatin1) (n = 10), and SCI + DHODH Inhibitor group (SCI + teriflunomide) (n = 10). Ten rats in the sham group only received laminectomy without SCI. To induce spinal cord injury, spinal cord injury surgery was performed in the middle thoracic region of rats (T8-T9).
Click to Show/Hide
|
||||
Response regulation | The application of DHODH is a potential treatment for spinal cord injury (SCI). DHODH can reduce the ferroptosis of neurons after spinal cord injury by regulating the P53/ALOX15 signaling pathway, thereby alleviating spinal cord injury. | ||||
Acute myeloid leukaemia [ICD-11: 2A60]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [35] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model |
hBMSCs (Bone marrow stromal cells) | ||||
K-562 cells | Chronic myelogenous leukemia | Homo sapiens | CVCL_0004 | ||
THP-1 cells | Childhood acute monocytic leukemia | Homo sapiens | CVCL_0006 | ||
HL-60 cells | Adult acute myeloid leukemia | Homo sapiens | CVCL_0002 | ||
MOLM-13 cells | Adult acute myeloid leukemia | Homo sapiens | CVCL_2119 | ||
In Vivo Model |
All BALB/C male nude mice (6 weeks old), purchased from Beijing Weitong lihua Experimental Animal Technology Co., Ltd, were maintained in pathogen-free facilities. The K562 or HL-60 cells (3 x 106, 200 ul) transfected with circKDM4C, vector, si-NC, or si-circKDM4C were subcutaneously injected into the right flank of nude mice (9 mice each group) for tumorigenesis. The maximum (Length) and minimum (Width) length of the tumors were measured.
Click to Show/Hide
|
||||
Response regulation | The overexpression of circKDM4C in acute myeloid leukemia (AML) cell lines inhibited the cell proliferation, migration, invasion, and promoted ferroptosis. The expression of circKDM4C and hsa-let-7b-5p are negatively correlated, while circKDM4C and p53 are positively correlated to AML patients. Moreover, circKDM4C induces ferroptosis by sponging hsa-let-7b-5p which upregulates the expression of P53. | ||||
Hepatocellular carcinoma [ICD-11: 2C12]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [39] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
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. | ||||
Cerebral ischemia [ICD-11: 8B10]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [42] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
Wild-type SD rats were kept in the Animal Experiment Center of Southeast University. Experimental rats were divided into 4 groups (n = 6 per group). The method of establishing the I/R model was provided in supplementary material. Then, we covered the ligation with gel. In order to fully cover the infarcted area of the heart, we chose to inject about 300 uL of mimics + Gel at 23 mm below the left atrial appendage (about the ligation). In order to prevent excessive irradiation of tissue burns, we selected each irradiation for 2 min to control the body surface temperature for a total of 10 min of irradiation.
Click to Show/Hide
|
||||
Response regulation | The mir-196c-3p mimic (mimics) and photothermal nanoparticles (BTN) were co-encapsulated in an injectable Gel (mimics + Gel/BTN) with NIR-II light-triggered release. Consequently, declined ferroptosis in cardiomyocytes and improved cardiac function, survival rate in rats was achieved through the controlled release of Gel/BTN mimics in ischemia-reperfusion (I/R) model to simultaneously inhibit ferroptosis hub genes NOX4, P53, and LOX expression. | ||||
Cerebral ischaemic stroke [ICD-11: 8B11]
In total 2 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [43] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
rRBMECs (Rat brain microvascular endothelial cells) | ||||
In Vivo Model |
Sprague Dawley (SD) rats (n = 60) aged three weeks were purchased from the Experimental Animal Center of Xiangya Hospital of Central South University. All rats were bred in a specific pathogen-free environment in 12-h lightdark cycle and fed with rodent diet and water. All rats were anaesthetized with inhaling isoflurane (2%, CAS NO. 64181101, Lunan Pharmaceutical Co., LTD. Shandong, China) and sacrificed by cervical dislocation. The whole brain was removed after opening the cranial cavity.
Click to Show/Hide
|
||||
Response regulation | OGD combined with hyperglycemic reperfusion promoted Meg3 expression and there was positive correlation between Meg3 and p53 expression in RBMVECs. Subsequently, p53 inhibited the activity of GPX4 by binding with its promoter. The Meg3- p53 signaling pathway mediated the ferroptosis of RBMVECs upon injury induced by OGD combined with hyperglycemic reperfusion and Meg3 has been considered as an important mediator in regulating diabetic brain ischemic injury. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [44] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
PC12 cells | Adrenal gland pheochromocytoma | Rattus norvegicus | CVCL_0481 | |
Response regulation | PVT1 regulates ferroptosis through miR-214-mediated p53 and TFR1. The discovery of PVT1 and miR-214 as potential targets for I/R also implies that PVT1 and miR-214 play critical roles in ferroptosis, shedding new light on the mechanism of ferroptosis in acute ischemic stroke. | ||||
Chronic heart failure [ICD-11: BD1Z]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [45] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Necroptosis | hsa04217 | ||||
NF-kappa B signaling pathway | hsa04064 | ||||
Cell Process | Cell ferroptosis | ||||
Cell pyroptosis | |||||
In Vitro Model |
rHTs (Rat hippocampal tissues) | ||||
In Vivo Model |
Male wild-type (WT) C57BL/6J mice aged 8 weeks were obtained from the Experimental Animal Center, Guangzhou University of Chinese Medicine. Sham and TAC mice received corresponding isotype i.p. injections. TAC + AAVMLK3- mice were generated by intravenous (i.v.) injection of adeno-associated viral vector-MLK3 vector (AAVMLK3-) (GenePharma, Shanghai, China) 14 and 21 days before TAC surgery. Sham + AAVNC and TAC + AAVNC mice received AAVNC i.v. injections. TAC + antagomir and TAC + agomir were generated by i.v. injection of antagomir and agomir (30 pmol/g) 14 and 21 days before TAC surgery, respectively.
Click to Show/Hide
|
||||
Response regulation | MLK3 (MAP3K11) mainly regulates the JNK/ p53 signaling pathway-mediated oxidative stress and that ferroptosis causes myocardial fibrosis in the advanced stages of chronic heart failure (CHF). Promoting the expression of miR-351 can inhibit the expression of MLK3, and significantly improve cardiac function in mice subjected to TAC. | ||||
Health [ICD-11: N.A.]
In total 2 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [46] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | Alumina | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hHCs (Hippocampal cells) | ||||
In Vivo Model |
Male healthy Wistar rats (six-week-old, provided by Experimental Animal Centre of Harbin Medical University, China) were used in this study. All rats (3-4 rats per cage) access to standard diet anddeionized waterad libitum and were placed in standard laboratory conditions. Seventy-two rats (weighing 200-220 g) were randomly divided into 4 groups (n = 18): AlNPs group was exposed to 50 mg/kg AlNPs (< 50nm, Sigma-Aldrich, USA) by gavage once a day for 90 days. CRS + AlNPs group was received CRS for 21 days and was exposed to 50 mg/kg AlNPs daily by gavage for 90 days. CRS + H2O group was subjected to CRS for 21 days and was given the same volume of deionized water daily by gavage for 90 days. The control (CON) group was given the same volume of deionized water daily and not affected by restraint stress for 90 days.
Click to Show/Hide
|
||||
Response regulation | Alumina nanoparticles (AlNPs) and CRS activated IFN-/ASK1/JNK (MAPK8) signaling pathway. Furthermore, IFN- neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. JNK inhibits ubiquitin-mediated p53 degradation by increasing phosphorylation of p53 at Ser6, which helps mediate oxidative stress to trigger ferroptosis. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response | [46] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Responsed Drug | R46A2 | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hHCs (Hippocampal cells) | ||||
In Vivo Model |
Male healthy Wistar rats (six-week-old, provided by Experimental Animal Centre of Harbin Medical University, China) were used in this study. All rats (3-4 rats per cage) access to standard diet anddeionized waterad libitum and were placed in standard laboratory conditions. Seventy-two rats (weighing 200-220 g) were randomly divided into 4 groups (n = 18): AlNPs group was exposed to 50 mg/kg AlNPs (< 50nm, Sigma-Aldrich, USA) by gavage once a day for 90 days. CRS + AlNPs group was received CRS for 21 days and was exposed to 50 mg/kg AlNPs daily by gavage for 90 days. CRS + H2O group was subjected to CRS for 21 days and was given the same volume of deionized water daily by gavage for 90 days. The control (CON) group was given the same volume of deionized water daily and not affected by restraint stress for 90 days.
Click to Show/Hide
|
||||
Response regulation | Alumina nanoparticles (AlNPs) and CRS activated IFN-/ASK1/JNK (MAPK8) signaling pathway. Furthermore, IFN- neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. JNK inhibits ubiquitin-mediated p53 degradation by increasing phosphorylation of p53 at Ser6, which helps mediate oxidative stress to trigger ferroptosis. | ||||
Traumatic brain injury [ICD-11: NA07]
In total 1 item(s) under this disease | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response | [47] | ||||
Target Regulator | Cellular tumor antigen p53 (TP53) | Protein coding | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hBCs (Brain cells) | ||||
In Vivo Model |
The Experimental Animal Center at Southern Medical University provided the wild-type (WT) adult male and female C57BL/6 mice (body weight, 22-25 g). Briefly, mice were anesthetized with an intraperitoneal injection of sodium pentobarbital (30 mg/kg), and mounted on a stereotaxic frame. A midsagittal incision was made in the scalp and a circular craniotomy (4.5 mm diameter) was made over the left parietotemporal cortex.
Click to Show/Hide
|
||||
Response regulation | P53-mediated ferroptosis contributes to the pathogenesis of traumatic brain injury (TBI). Furthermore, SIRT2 exerts a neuroprotective effect against TBI by suppressing p53-mediated ferroptosis. | ||||
Sevoflurane
[Approved]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [3] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Diamine acetyltransferase 1 (SAT1) | Driver | |||
Responsed Disease | Neurotoxicity | ICD-11: NE61 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hBCs (Brain cells) | ||||
In Vivo Model |
Pregnant rats were placed in a dedicated plastic chamber with ambient gas at a flow rate of 2L/min. Fer-1 solubilized in saline and 1% dimethyl sulfoxide (DMSO) and PD146176 (a specific 15LOX inhibitor) dissolved in corn oil containing 1% DMSO were administered intraperitoneally to rats at a dose of 5 mg/kg 1 h before each exposure, respectively. Similarly, 0.5 mg/kg Ku55933 (an ATM inhibitor), which is diluted in saline containing 1% DMSO, was intraperitoneally administered 2 h previously.
Click to Show/Hide
|
||||
Response regulation | Sevoflurane could enhance 15LO2-PEBP1 interaction and activate ATM and its downstream P53/SAT1 pathway, which might be attributed to excessive p-ATM nuclear translocation, indicating a potential therapeutic target for ameliorating sevoflurane-induced neurotoxicity. | ||||
Cyperquat
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [9] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | ||
Responsed Disease | Parkinson disease | ICD-11: 8A00 | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
PC12 cells | Adrenal gland pheochromocytoma | Rattus norvegicus | CVCL_0481 |
Response regulation | As a classic drug employed inin vitromodels of Parkinson's disease, 1-methyl-4-phenylpyridinium (MPP) can induce senescence in PC12 cells. The expression of the ferroptosis-related proteins ASCL4 was upregulated and FTH1 was downregulated, which promoted accumulation of lipid peroxides and eventually led to ferroptosis. By rescuing MPP-induced ferroptosis, cell senescence could be inhibited, and its molecular mechanism was related to a p53/SLC7A11/GPX4 signaling pathway. | |||
Tanshinone IIA
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [7] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | |||
Responsed Disease | Gastric cancer | ICD-11: 2B72 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
BGC-823 cells | Gastric carcinoma | Homo sapiens | CVCL_3360 | |
NCI-N87 cells | Gastric tubular adenocarcinoma | Homo sapiens | CVCL_1603 | ||
In Vivo Model |
All mice were housed under a setting of 12-h light/dark cycle at 22 ± 1, 55% humidity and fed with water and food provided at regular time. During the entire maintenance period, all mice were permitted free cage activity without joint immobilization. The initial body weights of the mice were between 20 and 23 grams. After subcutaneous injection of 2 x 106 BGC-823 gastric cancer cells into the back of NOD-SCID mice, the mice were treated with or without Tan IIA (50 mg/kg) or Tan IIA in combination with Fer-1 (50 mg/kg). Tan IIA was diluted in DMSO:Methanol:Hydroxypropyl-b-cydodextrin (HP-b-CD) = 1:1:1. Fer-1 was also dissolved in DMSO:Methanol:HP-b-CD. Seven days after BGC-823 gastric cancer cells injection, intraperitoneal injection with Tan IIA was carried out every other day followed by killing at day 22 of tumor cell inoculation. All mice were killed by dislocation of the cervical vertebrae. Before killing, the tumor volume was measured every 3 days. All experiments were carried out using six mice each group in three independent experiments of a time-dependent manner with three time points.
Click to Show/Hide
|
||||
Response regulation | Tanshinone IIA increased lipid peroxidation and up-regulated Ptgs2 and Chac1 expression, two markers of ferroptosis. In addition, Tan IIA also up-regulated p53 expression and down-regulated xCT (SLC7A11) expression. Therefore, Tan IIA could suppress the proliferation of gastric cancer via inducing p53 upregulation-mediated ferroptosis. | ||||
Seratrodast
[Discontinued in Phase 3]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [10] | ||||
Drug for Ferroptosis | Suppressor | ||||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | |||
Responsed Disease | Status epilepticus | ICD-11: 8A66 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
HT22 cells | Normal | Mus musculus | CVCL_0321 | |
In Vivo Model |
Drugs were dissolved in vehicle (0.1% DMSO + 20% PEG 300 + 0.5% CMC-Na + ddH2O). Mice in Control and PTZ groups were administered for five days with an equivalent volume of vehicle. PTZ-induced seizure model was done for the subsequent 1 h after the last administration of drugs. We performed a preliminary doseresponse trial, the dose of 60 mg/kg was established as being sufficient to trigger seizures with lower mortality and chosen as the optimal dose. One mouse in PTZ group was dead due to a severe seizure. At the end of the experiment, the mice were anesthetized or euthanized. For histopathological studies, the mice were anesthetized and intracardially perfused with 0.9% saline, followed by 0.4% paraformaldehyde for fixation of the brain. For immunoblot analysis, the hippocampus was rapidly isolated.
Click to Show/Hide
|
||||
Response regulation | Seratrodast could reduce lipid ROS production, regulate the system xc-/glutathione (GSH)/glutathione peroxidase 4 (GPX4) axis, and inhibit JNK (MAPK8) phosphorylation and p53 expression. JNK can directly or indirectly modulate the expression and activation of p53, which could regulate ferroptosis through inhibition of SLC7A11 transcription. Seratrodast increased the latency of seizures and reduced seizure duration in pentylenetetrazole-induced seizures in Epilepsy. | ||||
Apigenin
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [2] | ||||
Drug for Ferroptosis | Suppressor | ||||
Response Target | Phospholipid hydroperoxide glutathione peroxidase (GPX4) | Suppressor | |||
Responsed Disease | Status epilepticus | ICD-11: 8A66 | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
SH-SY5Y cells | Neuroblastoma | Homo sapiens | CVCL_0019 | |
In Vivo Model |
5-weeks-old kainate (KA)-induced BALB/c nude mice, a widely used epilepsy mouse model, were performed with intraperitoneal (i.p.) injection of KA (6 mg/kg). Pre-treatment 21 with antioxidant apigenin (60 mg/Kg, 2 days) or post-treatment with apigenin (60 mg/Kg, 1 day), mice were injected with KA (6 mg/kg) via intraperitoneal (i.p.) injection, and then HCP (0.5 mg/Kg) were injected by intravenous (i.v.) injection. In vivo and Ex vivo fluorescence images of relative ClO levels in mice brains 5, 15, 30, 45, and 60 min post injection of HCP were further performed by using the IVIS Spectrum imaging system (Nanjing University) with an excitation filter of 430 nm and the collection wavelength range is from 500-600 nm.
Click to Show/Hide
|
||||
Response regulation | Apigenin can efficiently reduce the expression of intracellular MPO and increase the levels of GPX4 and SIRT1, thereby conferring neuroprotection through regulation of kainic acid (KA)-induced ferroptosis. And the level of Ac-p53 inside the brains treated with apigenin was down-regulated, suggesting that the p53-mediated ferroptosis pathway might be blocked. Overall, apigenin was screened and confirmed as an efficient lead compound for epilepsy prevention and treatment. | ||||
Bavachin
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [6] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | ||
Responsed Disease | Osteosarcoma | ICD-11: 2B51 | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
MG-63 cells | Osteosarcoma | Homo sapiens | CVCL_0426 |
HOS cells | Osteosarcoma | Homo sapiens | CVCL_0312 | |
Response regulation | Bavachin could induce Osteosarcoma cell ferroptosis. Furthermore, bavachin elevated intracellular ferrous iron levels by increasing TFRC and DMT1 expression and decreasing FTH and FTL expressions. Bavachin also reduced SLC7A11 and GPX4 expression and promoted ROS and MDA accumulation by downregulating p-STAT3 to upregulate P53 expression. | |||
Ferric citrate
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [12] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | |||
Responsed Disease | Sarcopenia | ICD-11: FB32 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Glutathione metabolism | hsa00480 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
C2C12 cells | Normal | Mus musculus | CVCL_0188 | |
In Vivo Model |
The 8-week- and 40-week-old male SAMP8 mice were purchased from the model animal research center of Zhishan Institute of Healthcare Research Co., Ltd. (Beijing, China). All the mice were kept in an SPF grade animal facility at 24 with a relative humidity of 50%-60%, and in a light/dark cycle of 12 h/12 h.
Click to Show/Hide
|
||||
Response regulation | Ferric citrate induced ferroptosis in C2C12 cells, as well as impaired their differentiation from myoblasts to myotubes. Iron overload upregulated the expression of P53, which subsequently repressed the protein level of Slc7a11 (solute carrier family 7, member 11), a known ferroptosis-related gene. Targeting iron accumulation and ferroptosis might be a therapeutic strategy for treating sarcopenia. | ||||
Gambogenic Acid
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [8] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | ||
Responsed Disease | Melanoma | ICD-11: 2C30 | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell adhesion molecules | hsa04514 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
Cell migration | ||||
Cell invasion | ||||
In Vitro Model |
A-375 cells | Amelanotic melanoma | Homo sapiens | CVCL_0132 |
A2058 cells | Amelanotic melanoma | Homo sapiens | CVCL_1059 | |
Response regulation | Gambogenic acid (GNA) significantly inhibited the invasion, migration and EMT in melanoma cells, and these cells exhibited small mitochondrial wrinkling (an important feature of ferroptosis). GNA upregulated the expression of p53, solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) in the model cells, contributing to the mechanisms underlying GNA-induced ferroptosis. | |||
Isorhynchophylline
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [11] | ||||
Drug for Ferroptosis | Suppressor | ||||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | |||
Responsed Disease | Intracerebral hemorrhage | ICD-11: 8B00 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
p53 signaling pathway | hsa04115 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
HT22 cells | Normal | Mus musculus | CVCL_0321 | |
In Vivo Model |
Adult male Sprague-Dawley rats (SD rats, weighing 250-300 g) aged 11-12 weeks were purchased from SLAC Laboratory Animal Co., Ltd. (Shanghai, China). All 96 rats were randomly divided into four groups of 24 rats each: Sham group, Sham + IRN (30 mg/Kg) group, ICH group, and ICH + IRN (30 mg/Kg) group. The rats in sham group were injected with PBS solution, and the Sham + IRN (30 mg/Kg) group was received an equal amount of 30 mg/Kg IRN solution (intra-peritoneal injection) after the sham operation. After ICH, the rats in ICH group were injected with PBS solution, and the ICH + IRN (30 mg/Kg) group was received an equal amount of 30 mg/Kg IRN solution (intra-peritoneal injection).
Click to Show/Hide
|
||||
Response regulation | Isorhynchophylline (IRN) decreased ferroptosis and lipid ROS level, upregulated the expression of miR-122-5p and SLC7A11 mRNA, and inhibited TP53 expression. In conclusion, IRN protects neurocyte from intracerebral hemorrhage (ICH)-induced ferroptosis via miR-122-5p/TP53/SLC7A11 pathway, which may provide a potential therapeutic mechanism for ICH. | ||||
Kayadiol
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [5] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | ||
Responsed Disease | T-cell lymphoma | ICD-11: 2B01 | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
YT cells | Natural killer cell lymphoblastic leukemia | Homo sapiens | CVCL_1797 |
hPBLs (Human peripheral blood lymphocytes) | ||||
Response regulation | Kayadiol decreased the expression of SLC7A11 and GPX4, the negative regulatory proteins for ferroptosis. And p53 was the key mediator of kayadiol-induced ferroptosis by SLC7A11/GPX4 axis through p53 knockout experiments. Kayadiol can serve as an effective alternative in the treatment of NK/T cell lymphoma. | |||
Pseudolaric acid B
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [4] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Cystine/glutamate transporter (SLC7A11) | Driver; Suppressor | |||
Responsed Disease | Glioblastoma | ICD-11: 2A00 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
U-87MG cells | Glioblastoma | Homo sapiens | CVCL_0022 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
SHG-44 cells | Astrocytoma | Homo sapiens | CVCL_6728 | ||
In Vivo Model |
Twenty athymic BALB/c nude mice (aged 4 weeks, weight 20-22 g, from Shanghai laboratory animal Center, Shanghai, China) were housed in a specific pathogen-free environment. A total of 1 x 106 logarithmically growing C6 cells in 100 uL of PBS were subcutaneously injected into the right flank of each mouse. Therapeutic experiments were started when the tumor reached about 150 mm3 after about 7 days. The mice were allocated to receive intraperitoneal injections of vehicle (control group, n = 5/group), PAB at the dosage of 10 mg/kg body weight (n = 10/group) and 20 mg/kg body weight (n = 10/group) in the same volume 50 uL once a days for 8 times.
Click to Show/Hide
|
||||
Response regulation | Pseudolaric acid B (PAB) improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT (SLC7A11) pathway, which further exacerbated accumulation of H2O2and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment. | ||||
Quisinostat
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [1] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Phospholipid hydroperoxide glutathione peroxidase (GPX4) | Suppressor | |||
Responsed Disease | Oral squamous cell carcinoma | ICD-11: 2B6E | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Necroptosis | hsa04217 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell apoptosis | |||||
Cell pyroptosis | |||||
In Vitro Model |
CAL-27 cells | Tongue adenosquamous carcinom | Homo sapiens | CVCL_1107 | |
Tca8113 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6851 | ||
In Vivo Model |
Adult male athymic BALB/c nude mice (20-22 g of 5-week-old mice) were housed in a controlled environment at 23 ± 2 and 40%-70% humidity under a 12 h dark/light cycle with free access to irradiated food and sterile water. A suspension of 6 x 106/100 uL TCA-8113 cells was inoculated subcutaneously into the hind flank region of each nude mouse. The average tumor volume in nude mice reached 100 mm3, and mice were randomly divided into three groups. Quisinostat was formulated in normal saline and administered at 3 and 10 mg/kg/day byintraperitoneal injection. Control mice were given equal volume saline intraperitoneally. The tumor volume and the bodyweight of mice were monitored every three days.
Click to Show/Hide
|
||||
Response regulation | Quisinostat could increase the apoptosis rate in the tumor tissues of nude mice. Up-regulation of the expression of p53 and down-regulated expression of GPX4 in cell lines were observed by immunofluorescent staining, and the expression locations of p53 and GPX4 proteins in TSCC cells were observed. Quisinostat may be a potential drug for the treatment of tongue squamous cell carcinoma. | ||||
Levobupivacaine
[Approved]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [18] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Lung cancer | ICD-11: 2C25 | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model |
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | |
A427 cells | Lung carcinoma | Homo sapiens | CVCL_1055 | ||
In Vivo Model |
Balb/c nude mice (n= 5, 4-week-old, male) were applied to detect the impact of levobupivacaine on tumor growth. Mice subcutaneously injected with 1 x 107 A549 cells were treated with levobupivacaine (40 umol/Kg) or equal volume saline. The tumor volume was remarked every 5 days and finished at 30 days after injection, followed by the analysis of volume (length (width/2)2) and weight.
Click to Show/Hide
|
||||
Response regulation | Levobupivacaine could inhibit the proliferation and induce the apoptosis of non-small cell lung cancer (NSCLC) cells. Levobupivacaine was able to attenuate the invasion and migration in the cells. The treatment of levobupivacaine remarkably increased the levels of ROS, iron, and Fe2+ in NSCLC cells. Mechanically, levobupivacaine up-regulated the expression of p53 and induced ferroptosis by regulating p53 in NSCLC cells. | ||||
Flubendazole
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [20] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Prostate cancer | ICD-11: 2C82 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell apoptosis | |||||
In Vitro Model |
PC-3 cells | Prostate carcinoma | Homo sapiens | CVCL_0035 | |
DU145 cells | Prostate carcinoma | Homo sapiens | CVCL_0105 | ||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
RWPE-1 cells | Normal | Homo sapiens | CVCL_3791 | ||
In Vivo Model |
24 nude mice (3-4 weeks) were acquired from the experimental animal center of southern medical university (GuangZhou, China) and kept under specific pathogen-free conditions. 4 x 106 PC3 cells were implanted subcutaneously into the right armpit regions of each nude mouse. When the tumors volume reached approximately 40 mm3, mice were randomly divided into two groups to receive flubendazole (10 mg/kg, once daily), 5-fluorouracil (30 mg/kg, once daily), their combination, and vehicle control by intraperitoneal injection. After 20 days of treatment, all mice were sacrificed and tumor weight and tumor volume were immediately measured, respectively.
Click to Show/Hide
|
||||
Response regulation | Flubendazole is a novel P53 inducer which exerts anti-proliferation and pro-apoptosis effects in castration-resistant prostate cancer (CRPC) through hindering the cell cycle and activating the ferroptosis, and indicates that a novel utilization of flubendazole in neoadjuvant chemotherapy of CRPC. | ||||
Albiziabioside A
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [14] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Colon cancer | ICD-11: 2B90 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
MCF-7 cells | Breast carcinoma | Homo sapiens | CVCL_0031 | |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | ||
hBMECs (Human brain microvascular endothelial cells) | |||||
L-02 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_6926 | ||
HaCaT cells | Normal | Homo sapiens | CVCL_0038 | ||
HCT-8 cells | Ileocecal adenocarcinoma | Homo sapiens | CVCL_2478 | ||
Hep-G2 cells | Hepatoblastoma | Homo sapiens | CVCL_0027 | ||
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
CCD-841CoN cells | Normal | Homo sapiens | CVCL_2871 | ||
BEAS-2B cells | Normal | Homo sapiens | CVCL_0168 | ||
BEL-7402 cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_5492 | ||
In Vivo Model |
A total of 1 x 107 HCT116 cells were subcutaneously inoculated into the right flank of BALB/c mice. When tumor reached 70-100 mm3 (10 days after implant), mice were divided into five groups of eight animals at random. The groups with D13 were administered intravenously 20 mg/kg and 10 mg/kg. The positive control group was treated with AlbA(20 mg/kg and 10 mg/kg) through intravenous injection. The negative control group received 0.9% normal saline through intravenous injection.
Click to Show/Hide
|
||||
Response regulation | The study synthesized a series of Albiziabioside A derivatives and evaluated the antitumor activity both in vitro and in vivo. Compound D13 could induce apoptosis and ferroptosis through the mitochondrial pathway as a p53 activator. In addition, compound D13 significantly suppressed tumorigenesis without inducing toxicity in normal organs in vivo. The antitumor efficacy of D13 was further verified in colon cancer xenograft mouse models. | ||||
Apatinib
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [19] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Unspecific Target | |||
Responsed Disease | Ovarian cancer | ICD-11: 2C73 | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
Cell autophagy | ||||
In Vitro Model |
A2780 cells | Ovarian endometrioid adenocarcinoma | Homo sapiens | CVCL_0134 |
OVCAR-3 cells | Ovarian serous adenocarcinoma | Homo sapiens | CVCL_0465 | |
Response regulation | Apatinib combined with olaparib-induced ferroptosis via a p53-dependent manner in ovarian cancer. Further studies showed that apatinib combined with olaparib-induced ferroptosis by inhibiting the expression of Nrf2 and autophagy, thereby inhibiting the expression of GPX4. The Nrf2 activator RTA408 and the autophagy activator rapamycin rescued the combination drug-induced ferroptosis. | |||
Artesunate
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [21] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Unspecific Target | |||
Responsed Disease | Hereditary Leiomyomatosis | ICD-11: 2C90 | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model |
Caki-1 cells | Clear cell renal cell carcinoma | Homo sapiens | CVCL_0234 |
786-O cells | Renal cell carcinoma | Homo sapiens | CVCL_1051 | |
KTCTL-26 cells | Clear cell renal cell carcinoma | Homo sapiens | CVCL_5872 | |
A-498 cells | Renal cell carcinoma | Homo sapiens | CVCL_1056 | |
Response regulation | Artesunate (ART) significantly increased cytotoxicity and inhibited proliferation and clonogenic growth in both parental and sunitinib-resistant renal cell carcinoma (RCC) cells. P53 exclusively appeared in the KTCTL-26 cells, indicating that p53 might be predictive for ART-dependent ferroptosis. Thus, ART may hold promise for treating selected patients with advanced and even therapy-resistant RCC. | |||
BEBT-908
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [16] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Colorectal cancer | ICD-11: 2B91 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
PI3K-Akt signaling pathway | hsa04151 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
MC-38 cells | Colon adenocarcinoma | Homo sapiens | CVCL_B288 | |
In Vivo Model |
About 2 x 105 MC38 tumor cells were inoculated subcutaneously into C57BL/6 mice. A total of 100 mg/kg BEBT-908 was intravenously injected every other day for four times initiating from day 4 after tumor cell inoculation. Tumors were harvested on day 12 after inoculation, weighted, mechanically minced and incubated with 50 ug/mL DNase I (Sigma) and 2 mg/mL collagenase P (Sigma) for 20 minutes at 37.
Click to Show/Hide
|
||||
Response regulation | Treatment with BEBT-908 promoted ferroptotic cell death of cancer cells by hyperacetylating p53 and facilitating the expression of ferroptotic signaling. The dual PI3K/HDAC inhibitor BEBT-908 elicits potent antitumor responses, effectively inducing immunogenic ferroptosis of colorectal cancer cells and potentiating cancer immunotherapy. | ||||
Berberine
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [25] | ||||
Drug for Ferroptosis | Suppressor | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Cardiomyopathy | ICD-11: BC43 | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
All animal experiment protocols were implemented in accordance with the National Institutes of Health (NIH) guidelines, and the procedures were approved by the Animal Ethics Committee of Southwest University. C57BL/6J male mice, 8-10 weeks old, weighing 20 ± 2 g, were used in this study. Mice were housed under standard conditions at 22-24 with a 12 h light/12 h darkness cycle and free access to food and tap water. Thirty-six mice were randomly divided into six groups: control (N = 8), IMA group (50 mg/kg) (N = 8), Low-Ber (20 mg/kg) + IMA group (N = 8), Medium-Ber (40 mg kg1) + IMA group (N = 8), High-Ber (80 mg/kg) + IMA group (N = 8), and Fer-1 (1 mg/kg) + IMA group (N = 8). IMA was given intraperitoneally for 14 days. Ber was given orally 2 h before IMA treatment and Fer-1 was given intraperitoneally 2 h before IMA treatment.
Click to Show/Hide
|
||||
Response regulation | Berberine downregulated the expression of transferrin receptor (TfR) and P53 and upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), ferritin heavy chain-1 (FTH1), and glutathione peroxidase 4 (GPX4) in H9c2 cells and mice. The present data indicated that Ber has the potential to protect against IMA-induced cardiotoxicity, partlyviainhibiting Nrf2-dependent ferroptosis. | ||||
Clioquinol
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [23] | ||||
Drug for Ferroptosis | Suppressor | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Parkinson disease | ICD-11: 8A00 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
PI3K-Akt signaling pathway | hsa04151 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
SK-N-SH cells | Neuroblastoma | Homo sapiens | CVCL_0531 | |
In Vivo Model |
In total, twelve healthy adult rhesus monkeys (Macaca mulatta lasiotis, aged 4-5 years, and weighed 3.5-5 kg at the start of the study) were obtained from Sichuan Primed Biological Technology Co., Ltd. Monkeys were randomly divided into two groups: normal (control) group (n = 3) and MPTP group (n = 9). Monkeys from MPTP group were administered with MPTP by intramuscular injection daily at the beginning of the study, and then the MPTP dose was gradually added to 0.5 mg/kg at the end of the experiment. Monkeys from control group were injected with saline instead, and the other conditions were the same with MPTP group.
Click to Show/Hide
|
||||
Response regulation | Ferroptosis was probably involved in the pathogenesis of parkinson's disease (PD). Clioquinol (CQ) can decrease the excessive iron in the SN to normal level and directly protect DA neurons against oxidative stress probably by activating the AKT/mTOR survival pathway and blocking p53-medicated cell death. | ||||
Dihydroartemisinin
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [17] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Pancreatic cancer | ICD-11: 2C10 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model |
Panc02 cells | Pancreatic ductal adenocarcinoma | Mus musculus | CVCL_D627 | |
PANC-1 cells | Pancreatic ductal adenocarcinoma | Homo sapiens | CVCL_0480 | ||
In Vivo Model |
Six to eight-week-old female C57BL/6 mice were purchased from the Experimental Animal Center of Military Medical Sciences (Beijing, China). C57BL/6 mice were anesthetized and the tail of the pancreas was exposed. Panc 02 cells were resuspended in PBS at a concentration of 1 x 106 cells/0.1 ml and 50 ul cells were injected into the tail of the pancreas. Tumor-bearing mice were randomly divided into two groups (3 days after implantation). The control group was intraperitoneally injected 200 ul PBS daily for 10 days, and the DHA group was intraperitoneally injected with 100 mg/kg DHA daily for 10 days. The pancreatic tumors and spleens of the mice were collected for subsequent analysis.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin has anti-tumor effect in pancreatic cancer cells in vitro and in vivo. DHA treatment induced ferroptosis by increasing P53 and AOLX12 expression. | ||||
Ferric ammonium citrate
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [24] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Parkinson disease | ICD-11: 8A00 | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model |
MES23.5 cells | Normal | Mus musculus | CVCL_J351 | |
In Vivo Model |
Human a-synuclein (a-Syn) A53T overexpressiontransgenic mice(B6; C3-Tg (Prnp-SNCA*A53T) 83Vle/J) were originally obtained in breeding pairs from the Jackson Laboratory (004479) to generate a stable breeding colony. Animals were raised according to SPF level, kept at constant temperature (20 ± 2) , constant humidity (50 ± 10%), day and night cycle light (12-12 h), with free access to food and water.
Click to Show/Hide
|
||||
Response regulation | Ferroptosis firstly occurred in a relatively low concentration of ferric ammonium citrate (FAC)-treated group, and then apoptosis appeared in response to the increased iron doses. This was also confirmed in vivo in parkinson's disease transgenic mice and the underlying mechanism might be associated with the p53 signaling pathway, but not MAPK signaling pathway. | ||||
Ginkgo biflavones
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [15] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Colon cancer | ICD-11: 2B90 | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | |
HT29 cells | Colon cancer | Mus musculus | CVCL_A8EZ | ||
RKO cells | Colon carcinoma | Homo sapiens | CVCL_0504 | ||
LoVo cells | Colon adenocarcinoma | Homo sapiens | CVCL_0399 | ||
SW620 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0547 | ||
In Vivo Model |
HCT-116 cells (5 x 106) were injected into the flanks of 6-week-old male BALB/c nude mice to generate xenografts. The mice were randomly divided into four groups (n = 7 per group), and treatment was started at 96 h postinjection. The mice received an intraperitoneal injection (i.p.) of 0.9% saline solution containing 5% dimethyl sulfoxide (DMSO) (vehicle for blank control) and ginkgetin (10 mg/kg) once a day, respectively, and 5-FU (30 mg/kg) was used as the positive control (i.p., once every 3 days, alternately).
Click to Show/Hide
|
||||
Response regulation | Ginkgo biflavones can increase the expression level of p53 by inhibiting the expression of MDM2 protein and induce cell death independent of p53 transcriptional activity in vitro. And we provide evidence that ginkgetin strengthened the antitumor effect of fluorouracil (5-FU) in the HCT-116 colon cancer xenograft model. | ||||
Lipopolysaccharide
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [27] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Unspecific Target | |||
Responsed Disease | Lung injury | ICD-11: NB32 | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
MLE-12 cells | Normal | Mus musculus | CVCL_3751 |
Response regulation | Silence of MLK3 (MAP3K11) alleviated Lipopolysaccharide (LPS)-induced lung epithelial cell injury by inhibiting p53-mediated ferroptosis, suggesting that MLK3 may be a potential target to prevent acute lung injury. | |||
Methamphetamine
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [13] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Unspecific Target | |||
Responsed Disease | HIV Infection | ICD-11: 1C60 | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
BV-2 cells | Normal | Mus musculus | CVCL_0182 |
Response regulation | Methamphetamine (METH) and HIV-1 lead to oxidative stress and their combined effect increases the risk of HIV-associated neurocognitive disorder (HAND), which may be related to the synergistic ferroptotic impairment in microglia. We found that METH and HIV-1 Tat reduced the expression of ferroptotic protein GPX4 and the cell viability and enhanced the expression of P53 and the level of ferrous iron, while the above indices were significantly improved with pretreatment of ferrostatin-1. | |||
Olaparib
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [19] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Unspecific Target | |||
Responsed Disease | Ovarian cancer | ICD-11: 2C73 | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Cell Process | Cell ferroptosis | |||
Cell autophagy | ||||
In Vitro Model |
A2780 cells | Ovarian endometrioid adenocarcinoma | Homo sapiens | CVCL_0134 |
OVCAR-3 cells | Ovarian serous adenocarcinoma | Homo sapiens | CVCL_0465 | |
Response regulation | Apatinib combined with olaparib-induced ferroptosis via a p53-dependent manner in ovarian cancer. Further studies showed that apatinib combined with olaparib-induced ferroptosis by inhibiting the expression of Nrf2 and autophagy, thereby inhibiting the expression of GPX4. The Nrf2 activator RTA408 and the autophagy activator rapamycin rescued the combination drug-induced ferroptosis. | |||
Romidepsin
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [22] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Unspecific Target | |||
Responsed Disease | Adrenocortical carcinoma | ICD-11: 2D11 | ||
Pathway Response | Cell adhesion molecules | hsa04514 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model |
SW13 cells | Adrenal cortex carcinoma | Homo sapiens | CVCL_0542 |
Response regulation | HDAC inhibitor Romidepsin converted SW13+ cells also had reduced mRNA expression of the mitochondrial ROS detoxifier superoxide dismutase 2 (SOD2), and the tumor suppressor p53. HDAC inhibitor treatment synergistically increased adrenocortical carcinoma cell death following induction of ferroptosis. | |||
Zoledronic acid
[Investigative]
In total 1 item(s) under this drug | ||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [26] | |||
Drug for Ferroptosis | Inducer | |||
Response Target | Unspecific Target | |||
Responsed Disease | Osteonecrosis | ICD-11: FB81 | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model |
RAW 264.7 cells | Leukemia | Mus musculus | CVCL_0493 |
Response regulation | FBXO9 was downregulated in Zoledronic acid-treated osteoclast and promoted osteoclasts ferroptosis by inhibiting FBXO9-mediated p53 ubiquitination and degradation. The study provided a possible theoretical target for the clinical treatment of Bisphosphonates (BPs)-related osteonecrosis of jaw (BRONJ). | |||
Alumina
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [46] | ||||
Drug for Ferroptosis | Inducer | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Health | ICD-11: N.A. | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hHCs (Hippocampal cells) | ||||
In Vivo Model |
Male healthy Wistar rats (six-week-old, provided by Experimental Animal Centre of Harbin Medical University, China) were used in this study. All rats (3-4 rats per cage) access to standard diet anddeionized waterad libitum and were placed in standard laboratory conditions. Seventy-two rats (weighing 200-220 g) were randomly divided into 4 groups (n = 18): AlNPs group was exposed to 50 mg/kg AlNPs (< 50nm, Sigma-Aldrich, USA) by gavage once a day for 90 days. CRS + AlNPs group was received CRS for 21 days and was exposed to 50 mg/kg AlNPs daily by gavage for 90 days. CRS + H2O group was subjected to CRS for 21 days and was given the same volume of deionized water daily by gavage for 90 days. The control (CON) group was given the same volume of deionized water daily and not affected by restraint stress for 90 days.
Click to Show/Hide
|
||||
Response regulation | Alumina nanoparticles (AlNPs) and CRS activated IFN-/ASK1/JNK (MAPK8) signaling pathway. Furthermore, IFN- neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. JNK inhibits ubiquitin-mediated p53 degradation by increasing phosphorylation of p53 at Ser6, which helps mediate oxidative stress to trigger ferroptosis. | ||||
R46A2
[Investigative]
In total 1 item(s) under this drug | |||||
Experiment 1 Reporting the Ferroptosis-centered Drug Response | [46] | ||||
Drug for Ferroptosis | Suppressor | ||||
Response Target | Unspecific Target | ||||
Responsed Disease | Health | ICD-11: N.A. | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model |
hHCs (Hippocampal cells) | ||||
In Vivo Model |
Male healthy Wistar rats (six-week-old, provided by Experimental Animal Centre of Harbin Medical University, China) were used in this study. All rats (3-4 rats per cage) access to standard diet anddeionized waterad libitum and were placed in standard laboratory conditions. Seventy-two rats (weighing 200-220 g) were randomly divided into 4 groups (n = 18): AlNPs group was exposed to 50 mg/kg AlNPs (< 50nm, Sigma-Aldrich, USA) by gavage once a day for 90 days. CRS + AlNPs group was received CRS for 21 days and was exposed to 50 mg/kg AlNPs daily by gavage for 90 days. CRS + H2O group was subjected to CRS for 21 days and was given the same volume of deionized water daily by gavage for 90 days. The control (CON) group was given the same volume of deionized water daily and not affected by restraint stress for 90 days.
Click to Show/Hide
|
||||
Response regulation | Alumina nanoparticles (AlNPs) and CRS activated IFN-/ASK1/JNK (MAPK8) signaling pathway. Furthermore, IFN- neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. JNK inhibits ubiquitin-mediated p53 degradation by increasing phosphorylation of p53 at Ser6, which helps mediate oxidative stress to trigger ferroptosis. | ||||
References