Ferroptosis-centered Disease Response Information
General Information of the Disease (ID: DIS00158)
Name |
Health
|
||||
---|---|---|---|---|---|
ICD |
ICD-11: N.A.
|
Full List of Target(s) of This Ferroptosis-centered Disease
Phospholipid hydroperoxide glutathione peroxidase (GPX4)
In total 7 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Palmitic acid | Investigative | |||
Responsed Regulator | Heat shock factor protein 1 (HSF1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
In Vivo Model |
Hsf1 and Hsf1+/+-/- mice were kindly given as a present by Dr. Ivor J. Benjamin (Froedtert & Medical College of Wisconsin, Milwaukee, WI, USA). Sex-matched Hsf1-/- mice and Hsf1 littermates were used at 16-20 weeks old. Each mouse was injected intraperitoneally with 2.5 umol PA (dissolved in 0.5 mL 10% BSA) or an equal volume of BSA twice daily for 7 days.
Click to Show/Hide
|
||||
Response regulation | Palmitic acid (PA) decreased the protein expression levels of both heat shock factor 1 (HSF1) and glutathione peroxidase 4 (GPX4) in a dose- and time-dependent manner, which were restored by different ferroptosis inhibitors. Altogether, HSF1 may function as a key defender against PA-induced ferroptosis in cardiomyocytes by maintaining cellular iron homeostasis and GPX4 expression. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [10] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Selenium | Approved | |||
Pathway Response | Glutathione metabolism | hsa00480 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | mTCs (Mouse T cells) | ||||
In Vivo Model |
All mice used in this study were 6-12 weeks old on a C57/BL6/J background. WT or T-KO mice were fed with water supplemented with methionine (1 mgl-1, Sigma) or Se-Met (1 mgl-1, Sigma) and maintained on the diets for 4 weeks before experiments. Alternatively, WT mice were fed with selenium-adequate (0.15 mg/kg) and selenium-high (1 mg/kg) diets that were purchased from Envigo and mice were maintained on the diets for 4 weeks before experiments.
Click to Show/Hide
|
||||
Response regulation | The deletion of GPX4 in T cells selectively abrogated TFH cells and germinal center responses in immunized mice. Selenium supplementation enhanced GPX4 expression in T cells, increased TFH cell numbers and promoted antibody responses in immunized mice and young adults after influenza vaccination. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [11] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Alpha-Tocopherol | Investigative | |||
Pathway Response | Glutathione metabolism | hsa00480 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | mHSPCs (Mouse hematopoietic stem and progenitor cells) | ||||
In Vivo Model |
C57BL/6 WT mice were purchased from Beijing HFK BioScience Company (Beijing, China). Gpx4flox/flox mice were crossed with Vav-Cre mice and Mx-Cre mice to generate the Gpx4flox/flox Vav-Cre mice and Gpx4flox/flox Mx-Cre mice, respectively. For Gpx4 deletion, Gpx4flox/flox Mx-Cre mice were intraperitoneally injected with 20 mg/kg pIpC (Sigma) every other day for two weeks. CD45.1/45.2 mice and CD45.1 mice on a C57BL/6 background were used as competitor and recipient mice, respectively, in the competitive transplantation assay. Mice were fed natural ingredient diets containing >= 120 IU/kg vitamin E. A fixed formulation diet with or without 75 IU/kg vitamin E (Beijing HFK BioScience Company, Beijing, China) was fed to the mice involved in the vitamin E depletion experiments. For 5-FU treatment, mice were intraperitoneally injected with 150 mg/kg 5-FU (Sigma).
Click to Show/Hide
|
||||
Response regulation | a-Tocopherol, the main component of vitamin E, was shown to rescue the Gpx4-deficient hematopoietic stem and progenitor cells (HSPCs) from ferroptosis in vitro. When Gpx4 knockout mice were fed a vitamin E-depleted diet, a reduced number of HSPCs and impaired function of HSCs were found. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [12] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | D-2-hydroxyglutarate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
HT-1080 cells | Fibrosarcoma | Homo sapiens | CVCL_0317 | ||
KYSE-170 cells | Esophageal squamous cell carcinoma | Homo sapiens | CVCL_1358 | ||
Response regulation | Ectopic expression of mutant IDH1 or treatment of cells with cell-permeable D-2-hydroxyglutarate (D-2-HG) promotes the accumulation of lipid reactive oxygen species (ROS) and subsequently ferroptosis. Mechanistically, mutant IDH1 reduces the protein level of the glutathione peroxidase 4 (GPX4). | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [13] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Fluvastatin | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | Fluvastatin exerts potent protective effects against ox-LDL-induced endothelial cell dysfunction through regulation of GPx4 and xCT. These data indicated a novel function of fluvastatin in the protection of endothelial cells from ox-LDL-induced ferroptosis, the mechanism of which involves the regulation of GPx4 and xCT. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [14] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Ectonucleotide pyrophosphatase/phosphodiesterase family member 2 (ENPP2) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
In Vitro Model | CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
Response regulation | ENPP2 overexpression causes upregulation of GPX4 in H9c2 cells. In erastin-induced ferroptosis of H9c2 cells, both NRF2 and ACSL4 are increased, whereas ENPP2 overexpression reduces their expression in erastin-treated H9c2 cells. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [12] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Isocitrate dehydrogenase [NADP] cytoplasmic (IDH1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
HT-1080 cells | Fibrosarcoma | Homo sapiens | CVCL_0317 | ||
KYSE-170 cells | Esophageal squamous cell carcinoma | Homo sapiens | CVCL_1358 | ||
Response regulation | Ectopic expression of mutant IDH1 or treatment of cells with cell-permeable D-2-hydroxyglutarate (D-2-HG) promotes the accumulation of lipid reactive oxygen species (ROS) and subsequently ferroptosis. Mechanistically, mutant IDH1 reduces the protein level of the glutathione peroxidase 4 (GPX4). | ||||
Nuclear factor erythroid 2-related factor 2 (NFE2L2)
In total 6 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [2] | |||
Target for Ferroptosis | Marker/Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Drug | Calcitriol | Investigative | ||
Responsed Regulator | Kelch-like ECH-associated protein 1 (KEAP1) | Driver | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Pathways in cancer | hsa05200 | |||
NF-kappa B signaling pathway | hsa04064 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model | ZFL cells | Normal | Danio rerio | CVCL_3276 |
Response regulation | This study confirmed the protective effect of calcitriol on RSL3-induced ferroptosis in zebrafish liver cells, and reported for the first time that calcitriol inhibits ferroptosis in fish cells by regulating the Keap1/Nrf2/GPx4 axis and NF-kB/hepcidin axis. | |||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [15] | |||
Target for Ferroptosis | Marker/Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Drug | Berberine | Investigative | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model | HT22 cells | Normal | Mus musculus | CVCL_0321 |
Response regulation | Berberine can inhibit erastin-induced ferroptosis in HT22 cells possibly by activating the Nrf2-HO-1/ GPX4 pathway. | |||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [14] | |||
Target for Ferroptosis | Marker/Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Regulator | Ectonucleotide pyrophosphatase/phosphodiesterase family member 2 (ENPP2) | Suppressor | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
Cell migration | ||||
In Vitro Model | CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS |
Response regulation | ENPP2 overexpression causes upregulation of GPX4 in H9c2 cells. In erastin-induced ferroptosis of H9c2 cells, both NRF2 and ACSL4 are increased, whereas ENPP2 overexpression reduces their expression in erastin-treated H9c2 cells. | |||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [16] | |||
Target for Ferroptosis | Marker/Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Regulator | E3 ubiquitin-protein ligase SIAH2 (SIAH2) | Suppressor | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Ubiquitin mediated proteolysis | hsa04120 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 |
Phoenix-Eco cells | Normal | Homo sapiens | CVCL_H717 | |
i-MCF (Mouse primary cardiac fibroblasts) | ||||
Response regulation | NRF2 is a known SIAH2 target and master regulator of HO-1 expression. The increased vulnerability of SIAH2 knock-out cells to ferroptosis is probably due to several factors, including the increased expression of pro-ferroptotic HO-1 and the decreased expression of GPX4, a key factor for this iron-catalysed necrotic pathway. | |||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [17] | |||
Target for Ferroptosis | Marker/Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Regulator | Activin receptor type-1B (ACVR1B) | Driver | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Apoptosis | hsa04210 | |||
Necroptosis | hsa04217 | |||
Cell Process | Cell ferroptosis | |||
Cell apoptosis | ||||
Cell necrosis | ||||
In Vitro Model | HK-2 cells | Normal | Homo sapiens | CVCL_0302 |
Response regulation | The activin receptor-like kinase (ALK) 4/5 ( ACVR1B/TGFBR1), also known as activin-transforming growth factor (TGF) receptor, is involved in stress-induced renal injury. Pharmacological inhibition of ALK4/5 signaling attenuated erastin-induced ferroptosis by hyperactivating Nrf2 signaling in HK-2 cells. | |||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [17] | |||
Target for Ferroptosis | Marker/Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Regulator | TGF-beta receptor type-1 (TGFBR1) | Driver | ||
Pathway Response | Ferroptosis | hsa04216 | ||
Apoptosis | hsa04210 | |||
Necroptosis | hsa04217 | |||
Cell Process | Cell ferroptosis | |||
Cell apoptosis | ||||
Cell necrosis | ||||
In Vitro Model | HK-2 cells | Normal | Homo sapiens | CVCL_0302 |
Response regulation | The activin receptor-like kinase (ALK) 4/5 (ACVR1B/ TGFBR1), also known as activin-transforming growth factor (TGF) receptor, is involved in stress-induced renal injury. Pharmacological inhibition of ALK4/5 signaling attenuated erastin-induced ferroptosis by hyperactivating Nrf2 signaling in HK-2 cells. | |||
Heme oxygenase 1 (HMOX1)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [3] | |||
Target for Ferroptosis | Driver/Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Drug | SP600125 | Investigative | ||
Responsed Regulator | Mitogen-activated protein kinase 8 (MAPK8) | Driver | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model | BV-2 cells | Normal | Mus musculus | CVCL_0182 |
Response regulation | Following addition of the JNK (MAPK8) inhibitor SP600125, the expression of HO-1 decreased, expression of FTH1 was increased and iron accumulation was decreased. Therefore, it was hypothesized that NPs induced ferroptosis in BV2 cells via the JNK/HO-1/FTH1 pathway. | |||
Unspecific Target
In total 18 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [4] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | GW501516 | Discontinued in Phase 4 | |||
Responsed Regulator | Peroxisome proliferator-activated receptor delta (PPARD) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | mEFs (Mouse embryonic fibroblasts) | ||||
Response regulation | GW501516-activated PPARD stabilized peroxisomes through catalase upregulation by targeting peroxisomal hydrogen peroxide-mediated lysosomal rupture, which led to ferroptosis of xCT-deficient MEFs. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Alumina | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 8 (MAPK8) | Driver | |||
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 3 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Alumina | Investigative | |||
Responsed Regulator | Cellular tumor antigen p53 (TP53) | Driver | |||
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 4 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | R46A2 | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 8 (MAPK8) | Driver | |||
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 5 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | R46A2 | Investigative | |||
Responsed Regulator | Cellular tumor antigen p53 (TP53) | Driver | |||
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 6 Reporting the Ferroptosis-centered Disease Response by This Target | [23] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Acrolein | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Arabidopsis thalianasuspension cells (Arabidopsis thalianasuspension cells) | ||||
Response regulation | Acrolein, a lipid peroxide-derived reactive carbonyl species, is involved in plant ferroptosis-like cell death. The acrolein induced cell death could be mitigated by the known ferroptosis inhibitors such as Ferrostatin-1, Deferoxamine, a-Tocopherol, and glutathione. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [24] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Chebulagic Acid | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | rBMMSCs (Rat bone marrow mesenchymal stem cells) | ||||
In Vivo Model |
Sprague-Dawley rats (4 weeks) were obtained from the Animal Center of the Guangzhou University of Chinese Medicine. Procurement, maintenance, and treatment of the animals were performed under the supervision of the Institutional Animal Ethics Committee of the Guangzhou University of Chinese Medicine. The Gaussian 16 C.01 program was purchased from Guangzhou Molcalx Ltd. (Guangzhou, China).
Click to Show/Hide
|
||||
Response regulation | Two hydrolyzable tannins, chebulagic acid and chebulinic acid, can act as natural ferroptosis inhibitors. Their ferroptosis inhibition is mediated by regular antioxidant pathways (ROS scavenging and iron chelation), rather than the redox-based catalytic recycling pathway exhibited by Fer-1. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [24] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Chebulinic Acid | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | rBMMSCs (Rat bone marrow mesenchymal stem cells) | ||||
In Vivo Model |
Sprague-Dawley rats (4 weeks) were obtained from the Animal Center of the Guangzhou University of Chinese Medicine. Procurement, maintenance, and treatment of the animals were performed under the supervision of the Institutional Animal Ethics Committee of the Guangzhou University of Chinese Medicine. The Gaussian 16 C.01 program was purchased from Guangzhou Molcalx Ltd. (Guangzhou, China).
Click to Show/Hide
|
||||
Response regulation | Two hydrolyzable tannins, chebulagic acid and chebulinic acid, can act as natural ferroptosis inhibitors. Their ferroptosis inhibition is mediated by regular antioxidant pathways (ROS scavenging and iron chelation), rather than the redox-based catalytic recycling pathway exhibited by Fer-1. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [25] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | CyPPA | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Gluconeogenesis | hsa00010 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HT22 cells | Normal | Mus musculus | CVCL_0321 | |
In Vivo Model |
The C. elegans strain N2 was grown on NGM plates and on OP50 bacteria as a food source. For the lactate measurement, the nematodes were synchronized by bleaching with NaOH/NaClO and grown on NGM plates containing OP50 and either DMSO, 100 uM CyPPA or 250 uM CyPPA for 4 days. To determine lactate levels, the lactate assay kit was used (Sigma-Aldrich, MAK064). Briefly, worms were collected and washed to remove OP50. The worm pellet was snap-frozen, resuspended in lactate assay buffer and sonicated to break the cuticle. After centrifugation at 13000 rpm for 10 min, the supernatant was applied to a 10 kDa MWCO filter and centrifuged for 60 min at 13000 rpm to remove insoluble material.
Click to Show/Hide
|
||||
Response regulation | CyPPA rescued the neuronal cells from ferroptosis, while scavenging mitochondrial ROS and inhibiting glycolysis reduced its protection. Furthermore, SK channel activation increased survival of C. elegans challenged with mitochondrial toxins. | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [26] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Geraniin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | r-bmMSCs (Rat bone marrowderived mesenchymal stem cells) | ||||
Response regulation | Geraniin exhibits ferroptosis-inhibitory potential towards erastin-treated bmMSCs; such potential may mainly stem from its strong lipid peroxidation (LPO)-inhibition, Fe2+-chelating, and antioxidant actions. Geraniin gives neither dimer nor radical adduct, owing to the bulky HHDP (or DHHDP) group; thus, it is considered as a safe and effective ferroptosis-inhibitor. | ||||
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | 4-Methylumbelliferyl beta-D-xylopyranoside | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HT22 cells | Normal | Mus musculus | CVCL_0321 | |
Response regulation | The widely used sulfation inhibitor sodium chlorate and b-d-xyloside, which prevents proteoglycan glycosaminoglycan chain attachment, both reduced HS and CS, and exacerbated glutamate- and erastin-induced cell death, suggesting that extracellular matrix influenced oxytosis and ferroptosis. | ||||
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target | [28] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Butein | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | hBMSCs (Bone marrow stromal cells) | ||||
In Vivo Model |
Sprague-Dawley rats were purchased from the Animal Centre of Guangzhou University of Chinese Medicine. male Sprague-Dawley rats were collected, and the adherent soft tissues were removed. Both ends of the bones were cut away from the diaphysis with bone scissors. The bone marrow plugs were hydrostatically expelled from the bones by insertion of needles fastened to 10-mL syringes filled with complete medium; the needles were inserted into the distal ends of femora and proximal ends of the tibiae, and the marrow plugs expelled from the opposite ends.
Click to Show/Hide
|
||||
Response regulation | Butein and (S)-butin exert anti-ferroptotic action via an antioxidant pathway (especially the hydrogen atom transfer pathway). And butein displays superior antioxidant or anti-ferroptosis action to (S)-butin. | ||||
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target | [28] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Butin | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | hBMSCs (Bone marrow stromal cells) | ||||
In Vivo Model |
Sprague-Dawley rats were purchased from the Animal Centre of Guangzhou University of Chinese Medicine. male Sprague-Dawley rats were collected, and the adherent soft tissues were removed. Both ends of the bones were cut away from the diaphysis with bone scissors. The bone marrow plugs were hydrostatically expelled from the bones by insertion of needles fastened to 10-mL syringes filled with complete medium; the needles were inserted into the distal ends of femora and proximal ends of the tibiae, and the marrow plugs expelled from the opposite ends.
Click to Show/Hide
|
||||
Response regulation | Butein and (S)-butin exert anti-ferroptotic action via an antioxidant pathway (especially the hydrogen atom transfer pathway). And butein displays superior antioxidant or anti-ferroptosis action to (S)-butin. | ||||
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target | [29] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Salicylaldehyde isonicotinoyl hydrazone | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vivo Model |
A synchronous population was obtained by transferring egg-laying adults to fresh plates at 16 for 2-3 hr. The adults were removed and the plates with eggs then transferred to 25 to ensure sterility. After 48 hr at 25, when worms were at the late L4/young adult stage, 25-35 nematodes were transferred to fresh plates containing either vehicle control, 250 uM SIH, or 200 uM Lip-1. All plates were coded to allowing blinding of the experimenter to the treatment regime during scoring.
Click to Show/Hide
|
||||
Response regulation | Measures of lipid peroxidation and cell death show an increase with age and reduction by both Lip-1 and salicylaldehyde isonicotinoyl hydrazone (SIH) treatment. Blocking ferroptosis, either by inhibition of lipid peroxidation or by limiting iron retention, mitigates age-related cell death and markedly increases lifespan and healthspan. | ||||
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Sodium Chlorate | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HT22 cells | Normal | Mus musculus | CVCL_0321 | |
Response regulation | The widely used sulfation inhibitor sodium chlorate and b-d-xyloside, which prevents proteoglycan glycosaminoglycan chain attachment, both reduced HS and CS, and exacerbated glutamate- and erastin-induced cell death, suggesting that extracellular matrix influenced oxytosis and ferroptosis. | ||||
Experiment 16 Reporting the Ferroptosis-centered Disease Response by This Target | [30] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Ze 450 | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Gluconeogenesis | hsa00010 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HT22 cells | Normal | Mus musculus | CVCL_0321 | |
Response regulation | Ze 450 regulated HIF1 and cMyc protein levels in a shifted time dependency, which finally led to an enhanced glycolytic metabolism, by increasing HXKII and PDK1 protein levels. The protective effects against ferroptosis were mediated independently of estrogen receptor activation and were distinct from effects exerted by metformin. | ||||
Experiment 17 Reporting the Ferroptosis-centered Disease Response by This Target | [31] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Ethanolaminephosphotransferase 1 (Selenoi) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Response regulation | EPT1, also termed selenoprotein I (SELENOI), is speculated to directly synthesize phosphatidylethanolamine that drives ferroptosis. It also plays an indispensable role in myelination, neural development and maintaining phospholipid homeostasis in humans. This is consistent with our hypothesis that lowEPT1expression could induce AD through ferroptosis. | ||||
Experiment 18 Reporting the Ferroptosis-centered Disease Response by This Target | [32] | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Ferritin | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Response regulation | There is no excess ROS in discs with reducedwtsactivity, but ifFer1HCHis knocked-down simultaneously, very high ROS levels accumulate in the tissue, suggesting a protective role for the excess Fer1HCH inwtsmutants. | ||||
Polyunsaturated fatty acid lipoxygenase ALOX15B (ALOX15B)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [6] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Phosphatidylethanolamine-binding protein 1 (PEBP1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | hAECs (Human airway epithelial cells) | ||||
HT22 cells | Normal | Mus musculus | CVCL_0321 | ||
HK-2 cells | Normal | Homo sapiens | CVCL_0302 | ||
In Vivo Model |
Male 20 weeks old CD-1 mice were obtained from Charles River Laboratories. The mice were randomly assigned (using random number generator in Excel) to receive 5 mg/kg Fer-1 (Abcam, cat #ab146169) or 1.5% DMSO (vehicle) 60 minutes before injection of folic acid (Sigma-Aldrich) of 250 mg/kg in 0.3 mol/L sodium bicarbonate intraperitoneally. Mice were euthanized 48 h later.
Click to Show/Hide
|
||||
Response regulation | PEBP1, a scaffold protein inhibitor of protein kinase cascades, complexes with two 15LO isoforms, 15LO1 (ALOX15) and 15LO2 (ALOX15B) , and changes their substrate competence to generate hydroperoxy-PE. Inadequate reduction of hydroperoxy-PE due to insufficiency or dysfunction of a selenoperoxidase, GPX4, leads to ferroptosis. | ||||
Polyunsaturated fatty acid lipoxygenase ALOX15 (ALOX15)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [6] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Phosphatidylethanolamine-binding protein 1 (PEBP1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | hAECs (Human airway epithelial cells) | ||||
HT22 cells | Normal | Mus musculus | CVCL_0321 | ||
HK-2 cells | Normal | Homo sapiens | CVCL_0302 | ||
In Vivo Model |
Male 20 weeks old CD-1 mice were obtained from Charles River Laboratories. The mice were randomly assigned (using random number generator in Excel) to receive 5 mg/kg Fer-1 (Abcam, cat #ab146169) or 1.5% DMSO (vehicle) 60 minutes before injection of folic acid (Sigma-Aldrich) of 250 mg/kg in 0.3 mol/L sodium bicarbonate intraperitoneally. Mice were euthanized 48 h later.
Click to Show/Hide
|
||||
Response regulation | PEBP1, a scaffold protein inhibitor of protein kinase cascades, complexes with two 15LO isoforms, 15LO1 (ALOX15) and 15LO2 (ALOX15B) , and changes their substrate competence to generate hydroperoxy-PE. Inadequate reduction of hydroperoxy-PE due to insufficiency or dysfunction of a selenoperoxidase, GPX4, leads to ferroptosis. | ||||
Polyunsaturated fatty acid lipoxygenase ALOX12 (ALOX12)
In total 2 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [7] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | Hydrogen Sulfide | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | C2C12 cells | Normal | Mus musculus | CVCL_0188 | |
Response regulation | RSL3 induced ROS generation, inhibited CSE/H2S (Hydrogen sulfide) system, damaged mitochondrial structure, increased acetyl-CoA content and lipid peroxidation, which eventually lead to ferroptosis. Supplement of H2S normalized oxidative stress, acetyl-CoA content and ALOX12 acetylation and then protected myoblasts from RSL3-induced ferroptosis. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Histone-lysine N-methyltransferase 2D (KMT2D) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell differentiation | |||||
In Vitro Model | NHEK/SV3 cells | Normal | Homo sapiens | CVCL_9Q50 | |
3T3-J2 cells | Normal | Mus musculus | CVCL_W667 | ||
In Vivo Model |
All animal protocols were reviewed and approved by the Institutional Animal Care and Use Committee of the University of Pennsylvania. Mice were maintained on a mixed C57BL/6 background on a standard light-dark cycle. Mice carrying Mll4SET floxed alleles, Mll3SET floxed alleles, or a combination of both of these were crossed with Krt14-Cre transgenic mice.
Click to Show/Hide
|
||||
Response regulation | MLL4 (KMT2D) deficiency profoundly alters epidermal gene expression and uniquely rewires the expression of key genes and markers of ferroptosis (Alox12, Alox12b, and Aloxe3). | ||||
Polyunsaturated fatty acid 5-lipoxygenase (ALOX5)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [9] | |||
Target for Ferroptosis | Driver | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Drug | Zileuton | Approved | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model | HT22 cells | Normal | Mus musculus | CVCL_0321 |
Response regulation | Both the 5-LOX inhibitor zileuton and the ferropotosis inhibitor ferrostatin-1 acted through the same cascade to protect against glutamate oxidative toxicity. In conclusion, zileuton protected neurons from glutamate-induced oxidative stress at least in part by inhibiting ferroptosis. | |||
Long-chain-fatty-acid--CoA ligase 4 (ACSL4)
In total 10 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [14] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Ectonucleotide pyrophosphatase/phosphodiesterase family member 2 (ENPP2) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
In Vitro Model | CHO-S/H9C2 cells | Normal | Cricetulus griseus | CVCL_A0TS | |
Response regulation | ENPP2 overexpression causes upregulation of GPX4 in H9c2 cells. In erastin-induced ferroptosis of H9c2 cells, both NRF2 and ACSL4 are increased, whereas ENPP2 overexpression reduces their expression in erastin-treated H9c2 cells. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [18] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Proto-oncogene tyrosine-protein kinase Src (SRC) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | MCF-10A cells | Normal | Homo sapiens | CVCL_0598 | |
SUM159PT cells | Breast pleomorphic carcinoma | Homo sapiens | CVCL_5590 | ||
Hs-578T cells | Invasive breast carcinoma | Homo sapiens | CVCL_0332 | ||
In Vivo Model |
PDX models of triple-negative breast cancer were obtained from the Dana-Farber Cancer Institute and propagated in NSG mice. Tumors were harvested and digested using collagenase at 37. Once digested, the cells were filtered using a cell strainer (40 um), washed twice with PBS, and plated in DMEM/F12 (containing 10% FBS).
Click to Show/Hide
|
||||
Response regulation | 64-mediated activation of Src and STAT3 suppresses expression of ACSL4, an enzyme that enriches membranes with long polyunsaturated fatty acids and is required for ferroptosis. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [18] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Signal transducer and activator of transcription 3 (STAT3) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | MCF-10A cells | Normal | Homo sapiens | CVCL_0598 | |
SUM159PT cells | Breast pleomorphic carcinoma | Homo sapiens | CVCL_5590 | ||
Hs-578T cells | Invasive breast carcinoma | Homo sapiens | CVCL_0332 | ||
In Vivo Model |
PDX models of triple-negative breast cancer were obtained from the Dana-Farber Cancer Institute and propagated in NSG mice. Tumors were harvested and digested using collagenase at 37. Once digested, the cells were filtered using a cell strainer (40 um), washed twice with PBS, and plated in DMEM/F12 (containing 10% FBS).
Click to Show/Hide
|
||||
Response regulation | 64-mediated activation of Src and STAT3 suppresses expression of ACSL4, an enzyme that enriches membranes with long polyunsaturated fatty acids and is required for ferroptosis. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | hsa-mir-17 (Precursor RNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | The upregulated expression of individual miRNAs, miR-17, miR-18a, miR-19a, miR-20a, miR-19b and miR-92 were determined by qRT-PCR. This study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20-ACSL4 axis. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | hsa-mir-18a (Precursor RNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | The upregulated expression of individual miRNAs, miR-17, miR-18a, miR-19a, miR-20a, miR-19b and miR-92 were determined by qRT-PCR. This study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20-ACSL4 axis. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | hsa-mir-19a (Precursor RNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | The upregulated expression of individual miRNAs, miR-17, miR-18a, miR-19a, miR-20a, miR-19b and miR-92 were determined by qRT-PCR. This study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20-ACSL4 axis. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | hsa-mir-20a (Precursor RNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | The upregulated expression of individual miRNAs, miR-17, miR-18a, miR-19a, miR-20a, miR-19b and miR-92 were determined by qRT-PCR. This study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20-ACSL4 axis. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | hsa-miR-19b-3p (miRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | The upregulated expression of individual miRNAs, miR-17, miR-18a, miR-19a, miR-20a, miR-19b and miR-92 were determined by qRT-PCR. This study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20-ACSL4 axis. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | hsa-miR-92 (miRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | The upregulated expression of individual miRNAs, miR-17, miR-18a, miR-19a, miR-20a, miR-19b and miR-92 were determined by qRT-PCR. This study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20-ACSL4 axis. | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Tumor necrosis factor alpha-induced protein 3 (TNFAIP3) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HUVECs (Human umbilical vein endothelial cells) | ||||
Response regulation | The upregulated expression of individual miRNAs, miR-17, miR-18a, miR-19a, miR-20a, miR-19b and miR-92 were determined by qRT-PCR. This study revealed a novel mechanism through which miR-17-92 protects endothelial cells from erastin-induced ferroptosis by targeting the A20(TNFAIP3)-ACSL4 axis. | ||||
Hydroperoxide isomerase ALOXE3 (ALOXE3)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Histone-lysine N-methyltransferase 2D (KMT2D) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell differentiation | |||||
In Vitro Model | NHEK/SV3 cells | Normal | Homo sapiens | CVCL_9Q50 | |
3T3-J2 cells | Normal | Mus musculus | CVCL_W667 | ||
In Vivo Model |
All animal protocols were reviewed and approved by the Institutional Animal Care and Use Committee of the University of Pennsylvania. Mice were maintained on a mixed C57BL/6 background on a standard light-dark cycle. Mice carrying Mll4SET floxed alleles, Mll3SET floxed alleles, or a combination of both of these were crossed with Krt14-Cre transgenic mice.
Click to Show/Hide
|
||||
Response regulation | MLL4 (KMT2D) deficiency profoundly alters epidermal gene expression and uniquely rewires the expression of key genes and markers of ferroptosis (Alox12, Alox12b, and Aloxe3). | ||||
Ferritin light chain (FTL)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [20] | |||
Target for Ferroptosis | Suppressor | |||
Responsed Disease | Health [ICD-11: N.A.] | |||
Responsed Drug | JQ1 | Investigative | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
In Vitro Model | hDFs (Human dermal fibroblasts) | |||
Response regulation | JQ1 treatment reduced the expression of ferroptosis-resistance genes in senescent cells. And the treatment with JQ1 for 48 h showed decreased mRNA expressions of FTH and FTL. JQ1 treatment induced lipid peroxidation in senescent cells but not in non-senescent cells. | |||
Cystine/glutamate transporter (SLC7A11)
In total 3 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [21] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Drug | T-2 Toxin | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK-293T cells | Normal | Homo sapiens | CVCL_0063 | |
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
Response regulation | SLC7A11 overexpression significantly rescued the enhanced ferroptosis caused by T-2 toxin. T-2 toxin induces ferroptosis by downregulating SLC7A11 expression. Ferroptosis mediates T-2 toxin-induced cytotoxicity by increasing ROS and downregulating SLC7A11 expression. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [22] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | CD44 antigen (CD44) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK293 cells | Normal | Homo sapiens | CVCL_0045 | |
SK-N-BE(2)-C cells | Neuroblastoma | Homo sapiens | CVCL_0529 | ||
U2OS cells | Osteosarcoma | Homo sapiens | CVCL_0042 | ||
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | ||
SK-RC-42 cells | Renal cell carcinoma | Homo sapiens | CVCL_6192 | ||
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
T24 cells | Bladder carcinoma | Homo sapiens | CVCL_0554 | ||
UM-UC-3 cells | Bladder carcinoma | Homo sapiens | CVCL_1783 | ||
SW780 cells | Bladder carcinoma | Homo sapiens | CVCL_1728 | ||
In Vivo Model |
5.0 x 106 cells were mixed with Matrigel (BD Biosciences) at 1:1 ratio (v/v) and injected subcutaneously into seven-week old nude mice (NU/NU; Charles River). Mice were fed with regular chow. After nine weeks, the mice were killed and the tumors were weighed and recorded.
Click to Show/Hide
|
||||
Response regulation | Overexpression of the cancer stem cell marker CD44 enhanced the stability of SLC7A11 by promoting the interaction between SLC7A11 and OTUB1; depletion of CD44 partially abrogated this interaction. CD44 expression suppressed ferroptosis in cancer cells in an OTUB1-dependent manner. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [22] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Ubiquitin thioesterase OTUB1 (OTUB1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | HEK293 cells | Normal | Homo sapiens | CVCL_0045 | |
SK-N-BE(2)-C cells | Neuroblastoma | Homo sapiens | CVCL_0529 | ||
U2OS cells | Osteosarcoma | Homo sapiens | CVCL_0042 | ||
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | ||
SK-RC-42 cells | Renal cell carcinoma | Homo sapiens | CVCL_6192 | ||
NCI-H1299 cells | Lung large cell carcinoma | Homo sapiens | CVCL_0060 | ||
T24 cells | Bladder carcinoma | Homo sapiens | CVCL_0554 | ||
UM-UC-3 cells | Bladder carcinoma | Homo sapiens | CVCL_1783 | ||
SW780 cells | Bladder carcinoma | Homo sapiens | CVCL_1728 | ||
In Vivo Model |
5.0 x 106 cells were mixed with Matrigel (BD Biosciences) at 1:1 ratio (v/v) and injected subcutaneously into seven-week old nude mice (NU/NU; Charles River). Mice were fed with regular chow. After nine weeks, the mice were killed and the tumors were weighed and recorded.
Click to Show/Hide
|
||||
Response regulation | Overexpression of the cancer stem cell marker CD44 enhanced the stability of SLC7A11 by promoting the interaction between SLC7A11 and OTUB1; depletion of CD44 partially abrogated this interaction. CD44 expression suppressed ferroptosis in cancer cells in an OTUB1-dependent manner. | ||||
Arachidonate 12-lipoxygenase, 12R-type (ALOX12B)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Health [ICD-11: N.A.] | ||||
Responsed Regulator | Histone-lysine N-methyltransferase 2D (KMT2D) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell differentiation | |||||
In Vitro Model | NHEK/SV3 cells | Normal | Homo sapiens | CVCL_9Q50 | |
3T3-J2 cells | Normal | Mus musculus | CVCL_W667 | ||
In Vivo Model |
All animal protocols were reviewed and approved by the Institutional Animal Care and Use Committee of the University of Pennsylvania. Mice were maintained on a mixed C57BL/6 background on a standard light-dark cycle. Mice carrying Mll4SET floxed alleles, Mll3SET floxed alleles, or a combination of both of these were crossed with Krt14-Cre transgenic mice.
Click to Show/Hide
|
||||
Response regulation | MLL4 (KMT2D) deficiency profoundly alters epidermal gene expression and uniquely rewires the expression of key genes and markers of ferroptosis (Alox12, Alox12b, and Aloxe3). | ||||
References