General Information of the Disease (ID: DIS00114)
Name
Ischemia/reperfusion injury
ICD
ICD-11: DB98
Full List of Target(s) of This Ferroptosis-centered Disease
Phospholipid hydroperoxide glutathione peroxidase (GPX4)
In total 14 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Intestinal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Capsiate Investigative
Responsed Regulator Transient receptor potential cation channel subfamily V member 1 (TRPV1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model mSIOs (Mouse small intestinal organoids)
In Vivo Model
Six- to eight-week-old specific pathogen-free male C57BL/6 mice were purchased from the animal center of Nanfang Hospital of Southern Medical University (Guangzhou, China). The mice were anesthetized with isoflurane. A noninvasive microvascular artery clip was placed on the superior mesenteric artery (SMA) for 60 min, and the clip was removed for reperfusion for 2 hours. During the study period, body temperature was maintained at 37 with a heating pad, and liquid resuscitation was performed by subcutaneous injection with 0.5 ml of physiological saline immediately after reperfusion.

    Click to Show/Hide
Response regulation The gut microbiota metabolite capsiate enhances Gpx4 expression and inhibits ferroptosis by activating TRPV1 in intestinal ischemia/reperfusion (I/R) injury, providing a potential avenue for the management of intestinal ischemia/reperfusion (I/R) injury.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Gossypol acetic acid Investigative
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
A total of 55 adult male Sprague-Dawley rat (350-450 g) were anesthetized with urethane (1.5 g/kg, i.p.), then the hearts were perfused in a Langendorff system. After 30 min of stabilization, hearts were subjected to 30 min of global no-flow ischemia by stopping the perfusion. Reperfusion was followed with Krebs Henseleit (KH) buffer and GAA together for 2 h. A thermoregulated chamber kept the heart at 37 throughout the experiment. Control hearts were not subjected to I/R. The heart slices were sectioned at a thickness of 2 mm and stained with triphenyltetrazolium chloride (25 mg/100 mL) for 10 min and then fixed with 4% formaldehyde solution for 48 h to enhance color contrast.

    Click to Show/Hide
Response regulation Gossypol acetic acid significantly attenuated myocardial infarct size, reduced lipid peroxidation, decreased the mRNA levels of the ferroptosis markers Ptgs2 and Acsl4, decreased the protein levels of ACSL4 and NRF2, and increased the protein levels of GPX4 in I/R-induced ex vivo rat hearts. Thus, GAA may play a cytoprotectant role in ferroptosis-induced cardiomyocyte death and myocardial ischemia/reperfusion-induced ferroptotic cell death.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Target for Ferroptosis Suppressor
Responsed Disease Ischaemia reperfusion [ICD-11: DB98]
Responsed Drug Anhydrosafflor yellow B Investigative
Pathway Response Glutathione metabolism hsa00480
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell apoptosis
In Vitro Model PC12 cells Adrenal gland pheochromocytoma Rattus norvegicus CVCL_0481
Response regulation Hydroxysafflor yellow A (HSYA) and anhydrosafflor yellow B (AHSYB) limited ferroptosis and parthanatos to alleviate oxidative stress in PC12 cells. Oxygen glucose deprivation and reperfusion injury reduced GSH/GSSG level in PC12 cells, but the reduction of GSH/GSSG ratio was regained by HSYA or AHSYB. HSYA and AHSYB activated GPX4 and system Xc- to alleviate ferroptosis.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Baicalein Investigative
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
In this study, 30 male Sprague Dawley rats (325-375 g) anesthetized using pentobarbital (1.5 g/kg, i.p.) were used for heart infarct studies,Western blot analysis, and qPCR. The isolated hearts were perfused in a Langendorff system. A water-filled latex balloon was inserted into the left ventricle cavity via mitral valve and linked to a physiological pressure transducer (AD Instruments, MLT884) for continuous monitoring of left ventricular systolic pressure (LVSP) and end diastolic pressure (LVEDP). Left ventricular developed pressure (LVDP) was calculated as the difference between LVSP and LVEDP (LVDP = LVSP-LVEDP). Measurements were recorded using PowerLab system and Chart 8 software (ADInstrument, Bella Vista, New South Wales, Australia). The hearts were stable for 30 min, and then subjected to 45 min of global ischemia by halting perfusion, followed by 1 h of reperfusion with Krebs-Henseleit (KH) bicarbonate buffer gassed with 95% O2, 5% CO2 at 37 (pH 7.4). The infarcted myocardium was measured using triphenyltetrazolium chloride(TTC, 25 mg/mL) staining. The KH buffer containing 118 mM NaCl, 4.8 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 25 mM NaHCO 31.3 mM CaCl2, and 11 mM glucose was filtered through a 0.22 uM pore before use.

    Click to Show/Hide
Response regulation Baicalein and luteolin protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both baicalein and luteolin decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. Baicalein and luteolin reduced the ischemia/reperfusion-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Target for Ferroptosis Suppressor
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Drug Curcumin Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
rPTs (Rat pancreas tissues)
rHTs (Rat hippocampal tissues)
In Vivo Model
Forty female albino Wistar rats weighing 180-220 g were used in the study. Eight rats in each group were randomly assigned to five different groups: Group I (Sham); Group II (IR); Group III (IR + DMSO); Group IV (IR + Curcumin 100 mg/kg); and Group V (IR + 2 ug/kg LoxBlock-1) were determined. The animals were maintained at a temperature of 21 ± 2 and regulated humidity conditions (50 ± 5%) with a twelve-hour light/dark cycle. Throughout the experiment, the animals were fed standard commercial rat pellets and given tap water. All surgical and anesthesia procedures were performed understerile conditions. In addition, in a case of abnormal symptoms, the animals would be removed from the group and sacrificed under deep anesthesia.

    Click to Show/Hide
Response regulation Curcumin attenuates liver, pancreas and cardiac ferroptosis, oxidative stress and injury in ischemia/reperfusion-damaged rats by facilitating ACSL/GPx4 signaling.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Histochrome Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model mCMs (Mouse cardiomyocytes)
In Vivo Model
Male Fischer 344 rats (8 weeks old and 160 to 180 g; KOATECH, Pyeongtaek-si, Korea) were anesthetized by inhalation with 2% isoflurane and intubated using an 18-gauge intravenous catheter. The rats were mechanically ventilated with medical-grade oxygen. Surgery was performed on a 37 heating pad to prevent the body from getting cold. A left thoracotomy was performed after the chest was shaved to prevent contamination during surgery.

    Click to Show/Hide
Response regulation Histochrome treatment significantly increased GPx4 and free GSH levels, but decreased Cox-2 level. HC treatment significantly decreased intracellular and mitochondrial ROS levels by upregulating the expression of Nrf2 and antioxidant genes. The substantial cardioprotective effects of HC against myocardia I/R injury by reducing ferroptosis-associated myocardial injury.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Target for Ferroptosis Suppressor
Responsed Disease Ischaemia reperfusion [ICD-11: DB98]
Responsed Drug Hydroxysafflor Yellow A Investigative
Pathway Response Glutathione metabolism hsa00480
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell apoptosis
In Vitro Model PC12 cells Adrenal gland pheochromocytoma Rattus norvegicus CVCL_0481
Response regulation Hydroxysafflor yellow A (HSYA) and anhydrosafflor yellow B (AHSYB) limited ferroptosis and parthanatos to alleviate oxidative stress in PC12 cells. Oxygen glucose deprivation and reperfusion injury reduced GSH/GSSG level in PC12 cells, but the reduction of GSH/GSSG ratio was regained by HSYA or AHSYB. HSYA and AHSYB activated GPX4 and system Xc- to alleviate ferroptosis.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Luteolin Investigative
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
In this study, 30 male Sprague Dawley rats (325-375 g) anesthetized using pentobarbital (1.5 g/kg, i.p.) were used for heart infarct studies,Western blot analysis, and qPCR. The isolated hearts were perfused in a Langendorff system. A water-filled latex balloon was inserted into the left ventricle cavity via mitral valve and linked to a physiological pressure transducer (AD Instruments, MLT884) for continuous monitoring of left ventricular systolic pressure (LVSP) and end diastolic pressure (LVEDP). Left ventricular developed pressure (LVDP) was calculated as the difference between LVSP and LVEDP (LVDP = LVSP-LVEDP). Measurements were recorded using PowerLab system and Chart 8 software (ADInstrument, Bella Vista, New South Wales, Australia). The hearts were stable for 30 min, and then subjected to 45 min of global ischemia by halting perfusion, followed by 1 h of reperfusion with Krebs-Henseleit (KH) bicarbonate buffer gassed with 95% O2, 5% CO2 at 37 (pH 7.4). The infarcted myocardium was measured using triphenyltetrazolium chloride(TTC, 25 mg/mL) staining. The KH buffer containing 118 mM NaCl, 4.8 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 25 mM NaHCO 31.3 mM CaCl2, and 11 mM glucose was filtered through a 0.22 uM pore before use.

    Click to Show/Hide
Response regulation Baicalein and luteolin protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both baicalein and luteolin decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. Baicalein and luteolin reduced the ischemia/reperfusion-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [13]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator hsa-miR-135b-3p (miRNA) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
In Vivo Model
Male Sprague-Dawley rats aged 8-10 weeks and weighing 220 g were obtained from the Nanjing Biomedical Research Institute of Nanjing University. Following acclimatization for 1 week, the rats were divided into five groups of six rats each before the experiment. The establishment of the myocardial I/R model was based on previous studies . Sodium pentobarbital (45 mg/kg, i.p.) was used to anesthetize the rats, and the left coronary artery (LCA) was exposed using left thoracotomy at the fifth intercostal space. Following the LCA ligation with 7-0 silk sutures, a smooth catheter was applied to the artery to achieve ischemia for 30 min. The rats were then sacrificed 120 min after reperfusion. Rats in the sham group (without the LCA I/R) underwent surgery and were treated with saline. The miR-135b-3p group rats were injected with miR-135b-3p overexpression virus or knockdown lentivirus (1 x 108 U/ml, 0.2 ml), respectively, for five consecutive days before surgery.

    Click to Show/Hide
Response regulation MiR-135b-3p was found to promote the myocardial I/R injury by downregulating GPX4 expression. The results of this study elucidate a novel function of miR-135b-3p in exacerbating cardiomyocyte ferroptosis, providing a new therapeutic target for improving myocardial ischemia/reperfusion injury.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [14]
Target for Ferroptosis Suppressor
Responsed Disease Cardiac fibrosis [ICD-11: DB98]
Responsed Regulator rno-miR-375-3p (miRNA) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model rMTs (Rat myocardial tissues)
hCFs (Cardiac Fibroblasts)
In Vivo Model
Forty-two SD rats were randomly divided into sham operation group (n = 6) and I/R model group (n = 36). In the model group, rats were ligated with left anterior descending coronary artery to simulate MI. Specifically, after anesthetizing the animals in the I/R model group, an oblique incision was made in the third and fourth intercostal spaces of the left chest to expose the heart. Under a stereomicroscope, the junction of the pulmonary artery cone and the left atrial appendage was ligated with 6/0 noninvasive suture needle silk threads at 1-2 mm below the starting point of the coronary artery. Successful ischemia was indicated by ST segment elevation or T wave height and peaks of MI performance on electrocardiogram (ECG). The ligation was stopped after 45 minutes of ischemia, and the rats were maintained for 24 hours after reperfusion. As a drug treatment, 6 I/R model rats were treated with 20 nmol miRNA NC inhibitor (Thermo Fisher Scientific, Waltham, MA), 20 nmol miR-375-3p antagomir (Thermo Fisher Scientific, Waltham, MA) and 2 mg/kg Ferrostatin-1 (Fer-1; MCE, USA) for 28 days. The myocardial tissues of rats in the sham operation and I/R model groups as well as I/R model drug treatment group were then used for subsequent testing.

    Click to Show/Hide
Response regulation MiR-375-3p is an important factor inducing myocardial fibrosis after Myocardial infarction, which accelerates the ferroptosis of cardiomyocytes and promotes fibrosis by down-regulating GPX4.
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Suppressor
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator MIR9-3HG (IncRNA) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HL-1 cells Normal Mus musculus CVCL_0303
hBMSCs (Bone marrow stromal cells)
In Vivo Model
A total of 96 C57BL/6 male mice (20-25 g) aged 11-12 weeks were purchased from experimental animal center of experimental animal center of Guangdong Medical University. 96 mice were randomly divided into four groups (24 mice per group): Sham group (200 ul of PBS), Sham + BMSCs-Exo group (200 ul of BMSCs-Exo), I/R group (200 ul of PBS) and I/R + BMSCs-Exo group (200 ul of BMSCs-Exo). After 10 days of adaptive feeding, all mice were injected intraperitoneally with 0.4-0.5 mL/100 g 1%Pentobarbital Sodium. I/R and I/R + BMSCs-Exo group mice were subjected to cardiac I/R injury induced by ligation of the left anterior descending artery (LAD) for 30 min followed by 24 h reperfusion. Sham and Sham + BMSCs-Exo mice were sham treated and subjected to the same surgical procedures as I/R mice except that they did not receive ligation of the left anterior descending coronary artery.

    Click to Show/Hide
Response regulation Cellular ferroptosis is involved in the pathogenesis of Ischemia-Reperfusion Injury. BMSCs-Exo lncRNA Mir9-3hg can inhibit ferroptosis by modulating the Pum2/PRDX6 axis to exhibit cardioprotective effectsinvivoandinvitro. Silence of PRDX6 markedly decreased cell proliferation, GSH content and Gpx4 protein level, as well as prominently increased iron ion concentration and levels of ROS content and ACSL4 protein in H/R-treated HL-1 cells.
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Suppressor
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Pumilio homolog 2 (PUM2) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HL-1 cells Normal Mus musculus CVCL_0303
hBMSCs (Bone marrow stromal cells)
In Vivo Model
A total of 96 C57BL/6 male mice (20-25 g) aged 11-12 weeks were purchased from experimental animal center of experimental animal center of Guangdong Medical University. 96 mice were randomly divided into four groups (24 mice per group): Sham group (200 ul of PBS), Sham + BMSCs-Exo group (200 ul of BMSCs-Exo), I/R group (200 ul of PBS) and I/R + BMSCs-Exo group (200 ul of BMSCs-Exo). After 10 days of adaptive feeding, all mice were injected intraperitoneally with 0.4-0.5 mL/100 g 1%Pentobarbital Sodium. I/R and I/R + BMSCs-Exo group mice were subjected to cardiac I/R injury induced by ligation of the left anterior descending artery (LAD) for 30 min followed by 24 h reperfusion. Sham and Sham + BMSCs-Exo mice were sham treated and subjected to the same surgical procedures as I/R mice except that they did not receive ligation of the left anterior descending coronary artery.

    Click to Show/Hide
Response regulation Cellular ferroptosis is involved in the pathogenesis of Ischemia-Reperfusion Injury. BMSCs-Exo lncRNA Mir9-3hg can inhibit ferroptosis by modulating the Pum2/PRDX6 axis to exhibit cardioprotective effectsinvivoandinvitro. Silence of PRDX6 markedly decreased cell proliferation, GSH content and Gpx4 protein level, as well as prominently increased iron ion concentration and levels of ROS content and ACSL4 protein in H/R-treated HL-1 cells.
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Suppressor
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Peroxiredoxin-6 (PRDX6) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HL-1 cells Normal Mus musculus CVCL_0303
hBMSCs (Bone marrow stromal cells)
In Vivo Model
A total of 96 C57BL/6 male mice (20-25 g) aged 11-12 weeks were purchased from experimental animal center of experimental animal center of Guangdong Medical University. 96 mice were randomly divided into four groups (24 mice per group): Sham group (200 ul of PBS), Sham + BMSCs-Exo group (200 ul of BMSCs-Exo), I/R group (200 ul of PBS) and I/R + BMSCs-Exo group (200 ul of BMSCs-Exo). After 10 days of adaptive feeding, all mice were injected intraperitoneally with 0.4-0.5 mL/100 g 1%Pentobarbital Sodium. I/R and I/R + BMSCs-Exo group mice were subjected to cardiac I/R injury induced by ligation of the left anterior descending artery (LAD) for 30 min followed by 24 h reperfusion. Sham and Sham + BMSCs-Exo mice were sham treated and subjected to the same surgical procedures as I/R mice except that they did not receive ligation of the left anterior descending coronary artery.

    Click to Show/Hide
Response regulation Cellular ferroptosis is involved in the pathogenesis of Ischemia-Reperfusion Injury. BMSCs-Exo lncRNA Mir9-3hg can inhibit ferroptosis by modulating the Pum2/ PRDX6 axis to exhibit cardioprotective effectsinvivoandinvitro. Silence of PRDX6 markedly decreased cell proliferation, GSH content and Gpx4 protein level, as well as prominently increased iron ion concentration and levels of ROS content and ACSL4 protein in H/R-treated HL-1 cells.
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target [16]
Target for Ferroptosis Suppressor
Responsed Disease Cerebral ischemia/reperfusion [ICD-11: DB98]
Responsed Regulator BTB/POZ domain-containing adapter for CUL3-mediated RhoA degradation protein 2 (TNFAIP1) Driver
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Apoptosis hsa04210
Cell Process Cell ferroptosis
In Vitro Model PC12 cells Adrenal gland pheochromocytoma Rattus norvegicus CVCL_0481
Response regulation Downregulation of TNFAIP1 alleviates OGD/Rinduced neuronal damage by suppressing Nrf2/GPX4 mediated ferroptosis, which might lay the foundation for the investigation of targeted-therapy for cerebral ischemia-reperfusion injury in clinic.
Nuclear factor erythroid 2-related factor 2 (NFE2L2)
In total 9 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Myocardial ischemia/reperfusion [ICD-11: DB98]
Responsed Drug Micafungin Investigative
Responsed Regulator Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) Driver
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
The surgical procedure for establishing the myocardial I/R injury rat model was carried out as we did before. Briefly, a left thoracotomy was performed in the fourth intercostal space and the heart was exposed via opening thepericardium. The left coronary artery was surrounded with 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. At the end, the blood and hearts were collected for assay of the creatine kinase(CK) activity and infarct size to determine the success of I/R injury model. To explore the role of MALT1 in myocardial I/R injury the underlying mechanisms, three sets of experiment were performed.

    Click to Show/Hide
Response regulation The inhibition of MALT1 can reduce ischemia/reperfusion-induced myocardial ferroptosis through enhancing the Nrf2/SLC7A11 pathway; and MALT1 may be used as a potential target to seek novel or existing drugs (such as micafungin) for treating myocardial infarction.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [19]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Hepatic ischemia-reperfusion injury [ICD-11: DB98]
Responsed Drug Dimethyl fumarate Approved
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model AML12 cells Normal Mus musculus CVCL_0140
In Vivo Model
The mice were randomly divided into four groups of six: sham + vehicle, sham + DMF, IR + vehicle, and IR + DMF. The mice were supplemented with DMF at a concentration of 100 mg/kg or DMSO by daily oral gavage for a week before surgery, as previously reported. As stated in a prior study, the partial warm liver IRI model was developed. Briefly, the sham group only had free hepatic portal blood vessels after laparotomy, and the blood flow was not obstructed. As for the hepatic IR group, the blood supply to the left and mid-hepatic lobes was blocked, resulting in 70% mouse liver IRI for 90 min. The mice were put on a heated blanket after surgery in order to maintain body temperature and monitor vital signs. Blood supply was restored for 6 h. Died mice were eliminated for testing prior to sample collection. The mice were euthanized after the sample were obtained. The same experimenter carried out all surgeries.

    Click to Show/Hide
Response regulation NRF2 knockdown notably decreased the expression of SLC7A11 and HO-1 and blocked the anti-ferroptosis effects of dimethyl fumarate (DMF). DMF inhibits ferroptosis by activating the NRF2/SLC7A11/HO-1 axis and exerts a protective effect against hepatic ischemia-reperfusion injury.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Gossypol acetic acid Investigative
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
A total of 55 adult male Sprague-Dawley rat (350-450 g) were anesthetized with urethane (1.5 g/kg, i.p.), then the hearts were perfused in a Langendorff system. After 30 min of stabilization, hearts were subjected to 30 min of global no-flow ischemia by stopping the perfusion. Reperfusion was followed with Krebs Henseleit (KH) buffer and GAA together for 2 h. A thermoregulated chamber kept the heart at 37 throughout the experiment. Control hearts were not subjected to I/R. The heart slices were sectioned at a thickness of 2 mm and stained with triphenyltetrazolium chloride (25 mg/100 mL) for 10 min and then fixed with 4% formaldehyde solution for 48 h to enhance color contrast.

    Click to Show/Hide
Response regulation Gossypol acetic acid significantly attenuated myocardial infarct size, reduced lipid peroxidation, decreased the mRNA levels of the ferroptosis markers Ptgs2 and Acsl4, decreased the protein levels of ACSL4 and NRF2, and increased the protein levels of GPX4 in I/R-induced ex vivo rat hearts. Thus, GAA may play a cytoprotectant role in ferroptosis-induced cardiomyocyte death and myocardial ischemia/reperfusion-induced ferroptotic cell death.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [20]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Etomidate Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model rMTs (Rat myocardial tissues)
In Vivo Model
Male Sprague-Dawley rats (8 weeks, 180 g-210 g) were provided by the experimental animal center of Beijing Institute of Life Sciences. Rats were anesthetized with 2.5% sodium pentobarbital and fixed in supine position. The LAD was ligated using a 6-0 silk for a 30 min ischemic period. Ischemia was confirmed by discoloration of heart surface and ST elevation on the electrocardiogram (ECG) recording. After 30 min, the LAD ligation was released and the reperfusion was continued for 3 h, which was confirmed by the redness of the heart surface and the decrease in ST recorded by ECG. Rats in Sham group (n = 12) underwent the same surgical procedures, except that LAD was threaded but not ligated.

    Click to Show/Hide
Response regulation Etomidate (Eto) attenuated MIRI-induced heart failure, pathological damage, myocardial fibrosis, andinflammation, which may be related to its inhibition onferroptosis. Mechanically, the protection of Eto in myocardial ischemia reperfusion (MIR) injury (MIRI) may be achieved by activating Nrf2 pathway.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Histochrome Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model mCMs (Mouse cardiomyocytes)
In Vivo Model
Male Fischer 344 rats (8 weeks old and 160 to 180 g; KOATECH, Pyeongtaek-si, Korea) were anesthetized by inhalation with 2% isoflurane and intubated using an 18-gauge intravenous catheter. The rats were mechanically ventilated with medical-grade oxygen. Surgery was performed on a 37 heating pad to prevent the body from getting cold. A left thoracotomy was performed after the chest was shaved to prevent contamination during surgery.

    Click to Show/Hide
Response regulation Histochrome treatment significantly increased GPx4 and free GSH levels, but decreased Cox-2 level. HC treatment significantly decreased intracellular and mitochondrial ROS levels by upregulating the expression of Nrf2 and antioxidant genes. The substantial cardioprotective effects of HC against myocardia I/R injury by reducing ferroptosis-associated myocardial injury.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [21]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Lung ischemia/reperfusion Injury [ICD-11: DB98]
Responsed Drug Iridin Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model MLE-12 cells Normal Mus musculus CVCL_3751
In Vivo Model
In vivo, the LIRI model was established as described earlier. All mice were anesthetized with pentobarbital administered intraperitoneally (50 mg/kg, Sigma-Aldrich, MO, USA). After endotracheal intubation, the mice were ventilated using a rodent ventilator (MiniVent, Harvard Apparatus, USA), with the title volume set to 7 ml/kg, the respiratory rate set to 120 times/min, and the inspiratory/expiratory ratio set to 1: 2. A noninvasive clamp was used to interrupt the left pulmonary hilum, causing lung ischemia. The clamp was released after 60 minutes of ischemia, and the left lung was reperfused for 120 minutes. Animals were euthanized via cervical dislocation at the end of the experiment. Following that, lung specimens and bronchoalveolar lavage fluid were harvested for analysis. All procedures except lung ischemia were performed on mice in the sham group.

    Click to Show/Hide
Response regulation As a result, irisin postconditioning may protect against lung I/R damage by suppressing ferroptosis via the Nrf2/HO-1 signaling axis.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [22]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Naringenin Investigative
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
20 Sprague Dawley (SD) rats (6-8 weeks, 200-220 g) were acquired from the Second Clinical College of Guangzhou University of Traditional Chinese Medicine, and were weighed, coded, and randomly assigned to experimental groups. Rats were divided into Sham group, MI/R group, MI/R +NAR (low dose, 10 mg/kg/d) group, and MI/R +NAR (high dose, 50 mg/kg/d) group. For MI/R model, rats were anaesthetized by intraperitoneal injection of 1% pentobarbital sodium (60 mg/kg) and then received mechanical ventilation from an animal ventilator after endotracheal intubation.

    Click to Show/Hide
Response regulation Naringenin alleviated MI/R-induced pathological damage, inflammation and lipid peroxidation in myocardial tissue of rats. NAR adjusted the NRF2 /System xc - /GPX4 axis and improved ferroptosis. In conclusion, NAR can alleviate myocardial ischemia-reperfusion injury by regulating the Nrf2/System xc-/GPX4 axis to inhibit ferroptosis.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [23]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Lung ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Salidroside Investigative
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model MLE-12 cells Normal Mus musculus CVCL_3751
RAW 264.7 cells Leukemia Mus musculus CVCL_0493
In Vivo Model
Following endotracheal intubation, mice were ventilated with room air at a rate of 120 cycles/min and atidal volumeof 7 mL/kg (MiniVent, Harvard Apparatus, USA). To induce ischemia, mice underwent left thoracotomy, and the left pulmonary hilum was blocked for 60 min with a microvascular clamp. After ischemia, the coronary artery was reperfused for 120 min by removing the clamp. The mice were euthanized at the end of the experiment through CO2 asphyxiation and cervical dislocation. Next, bronchoalveolar lavage fluid (BALF), blood, and lung samples were collected for testing.

    Click to Show/Hide
Response regulation Salidroside postconditioning attenuates ferroptosis-mediated lung ischemia-reperfusion injury by activating the Nrf2/SLC7A11 signaling axis.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [16]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Cerebral ischemia/reperfusion [ICD-11: DB98]
Responsed Regulator BTB/POZ domain-containing adapter for CUL3-mediated RhoA degradation protein 2 (TNFAIP1) Driver
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Apoptosis hsa04210
Cell Process Cell ferroptosis
In Vitro Model PC12 cells Adrenal gland pheochromocytoma Rattus norvegicus CVCL_0481
Response regulation Downregulation of TNFAIP1 alleviates OGD/Rinduced neuronal damage by suppressing Nrf2/GPX4 mediated ferroptosis, which might lay the foundation for the investigation of targeted-therapy for cerebral ischemia-reperfusion injury in clinic.
Cystine/glutamate transporter (SLC7A11)
In total 6 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion [ICD-11: DB98]
Responsed Drug Micafungin Investigative
Responsed Regulator Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) Driver
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
The surgical procedure for establishing the myocardial I/R injury rat model was carried out as we did before. Briefly, a left thoracotomy was performed in the fourth intercostal space and the heart was exposed via opening thepericardium. The left coronary artery was surrounded with 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. At the end, the blood and hearts were collected for assay of the creatine kinase(CK) activity and infarct size to determine the success of I/R injury model. To explore the role of MALT1 in myocardial I/R injury the underlying mechanisms, three sets of experiment were performed.

    Click to Show/Hide
Response regulation The inhibition of MALT1 can reduce ischemia/reperfusion-induced myocardial ferroptosis through enhancing the Nrf2/SLC7A11 pathway; and MALT1 may be used as a potential target to seek novel or existing drugs (such as micafungin) for treating myocardial infarction.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [19]
Target for Ferroptosis Suppressor
Responsed Disease Hepatic ischemia-reperfusion injury [ICD-11: DB98]
Responsed Drug Dimethyl fumarate Approved
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model AML12 cells Normal Mus musculus CVCL_0140
In Vivo Model
The mice were randomly divided into four groups of six: sham + vehicle, sham + DMF, IR + vehicle, and IR + DMF. The mice were supplemented with DMF at a concentration of 100 mg/kg or DMSO by daily oral gavage for a week before surgery, as previously reported. As stated in a prior study, the partial warm liver IRI model was developed. Briefly, the sham group only had free hepatic portal blood vessels after laparotomy, and the blood flow was not obstructed. As for the hepatic IR group, the blood supply to the left and mid-hepatic lobes was blocked, resulting in 70% mouse liver IRI for 90 min. The mice were put on a heated blanket after surgery in order to maintain body temperature and monitor vital signs. Blood supply was restored for 6 h. Died mice were eliminated for testing prior to sample collection. The mice were euthanized after the sample were obtained. The same experimenter carried out all surgeries.

    Click to Show/Hide
Response regulation NRF2 knockdown notably decreased the expression of SLC7A11 and HO-1 and blocked the anti-ferroptosis effects of dimethyl fumarate (DMF). DMF inhibits ferroptosis by activating the NRF2/SLC7A11/HO-1 axis and exerts a protective effect against hepatic ischemia-reperfusion injury.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [23]
Target for Ferroptosis Suppressor
Responsed Disease Lung ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Salidroside Investigative
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model MLE-12 cells Normal Mus musculus CVCL_3751
RAW 264.7 cells Leukemia Mus musculus CVCL_0493
In Vivo Model
Following endotracheal intubation, mice were ventilated with room air at a rate of 120 cycles/min and atidal volumeof 7 mL/kg (MiniVent, Harvard Apparatus, USA). To induce ischemia, mice underwent left thoracotomy, and the left pulmonary hilum was blocked for 60 min with a microvascular clamp. After ischemia, the coronary artery was reperfused for 120 min by removing the clamp. The mice were euthanized at the end of the experiment through CO2 asphyxiation and cervical dislocation. Next, bronchoalveolar lavage fluid (BALF), blood, and lung samples were collected for testing.

    Click to Show/Hide
Response regulation Salidroside postconditioning attenuates ferroptosis-mediated lung ischemia-reperfusion injury by activating the Nrf2/SLC7A11 signaling axis.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [31]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator SEMA5A-IT1 (IncRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model AC16 [Human hybrid cardiomyocyte] cells Normal Homo sapiens CVCL_4U18
Response regulation SEMA5A-IT1 overexpression upregulated the expression of BCL2 and SLC7A11 through sponging miR-143-3p, thereby protecting cardiomyocytes against apoptotic and ferroptosis cell death. In conclusion, we propose that SEMA5A-IT1, which is transported to cardiomyocytes through circulating sEVs, is an important regulatory molecule that protects cardiomyocytes from ischemia-reperfusion injury.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [31]
Target for Ferroptosis Suppressor
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator hsa-miR-143-3p (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model AC16 [Human hybrid cardiomyocyte] cells Normal Homo sapiens CVCL_4U18
Response regulation SEMA5A-IT1 overexpression upregulated the expression of BCL2 and SLC7A11 through sponging miR-143-3p, thereby protecting cardiomyocytes against apoptotic and ferroptosis cell death. In conclusion, we propose that SEMA5A-IT1, which is transported to cardiomyocytes through circulating sEVs, is an important regulatory molecule that protects cardiomyocytes from ischemia-reperfusion injury.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [32]
Target for Ferroptosis Suppressor
Responsed Disease Cerebral ischemia/reperfusion [ICD-11: DB98]
Responsed Regulator hsa-miR-27a-3p (miRNA) Driver
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model hBCs (Brain cells)
In Vivo Model
The rats were anesthetized by intraperitoneal injection of pentobarbital sodium (40 mg/kg). Before the experiment, the rats were fasted for 8-10 h and ensured sufficient water. The left common carotid artery (CCA) and left external carotid artery (ECA) were ligated after skin incision in the middle of the neck. In the common carotid artery cut a small incision. In order to induce ischemia, a cord plug was inserted through the incision into the internal carotid artery (ICA) and then to the origin of the middle cerebral artery (MCA). After ischemia for 2 h, the rats were anesthetized and gently pulled out the cord plug. At the end of the reperfusion time, the rats were euthanized by injection of a minimal lethal dose of anesthesia.

    Click to Show/Hide
Response regulation The results of dual luciferase reporter gene technique indicated SLC7A11 as the target gene of miR-27a. In the current study, miR-27a upregulates ferroptosis to aggravate cerebral ischemia-reperfusion injury by SLC7A11.
Unspecific Target
In total 10 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [3]
Responsed Disease Intestinal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Apigenin Investigative
Responsed Regulator Amine oxidase [flavin-containing] B (MAOB) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HUVECs (Human umbilical vein endothelial cells)
In Vivo Model
Mice were exposed to 12/12 h of light/darkness and given free access to food and water. All C57BL/6J mice were fasted for 12 h and anesthetized by intraperitoneal injection of 1% sodium pentobarbital before modeling. Later, the superior mesenteric artery (SMA) was subjected to 45 min of global no-flow ischemia, followed by 90 min of reperfusion to induce IIRI. The successful model could be revealed by the microcirculation detector. In order to investigate the effects of APG, mice were randomly divided into different groups: Sham group, IIRI group, APG groups (2 mg/kg, 4 mg/kg and 8 mg/kg), ZnPPIX group (Protoporphyrin Zinc(), Dalian Meilun Biotech Co., Ltd., China, 10mg/kg), Selegiline group (10 mg/kg, Shandong Topscience Biotech Co., Ltd., China) and the ZnPPIX + Selegiline group (10 mg/kg ZnPPIX and 10 mg/kg Selegiline). As mentioned above, drug was intraperitoneal injected after a 10-min-ischemia.

    Click to Show/Hide
Response regulation MST analysis suggested that Apigenin could specifically bind to heme oxygenase 1 (HO-1) and monoamine oxidase b (MAO-B). Simultaneously, APG could attenuate ROS generation and Fe2+ accumulation, maintain mitochondria function thus inhibit ferroptosis and alleviate intestinal ischemia-reperfusion injury.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [4]
Responsed Disease Lung ischemia/reperfusion Injury [ICD-11: DB98]
Responsed Drug Lidocaine Investigative
Responsed Regulator Mitogen-activated protein kinase 14 (MAPK14) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
In Vitro Model A-549 cells Lung adenocarcinoma Homo sapiens CVCL_0023
Response regulation Lidocaine could regulate inflammation, oxidative stress and ferroptosis by blocking the p38 MAPK signaling pathway. Thus, lidocaine could act as a novel therapeutic treatment of patients with Lung Ischemia-reperfusion (I/R) injury.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [33]
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Deferoxamine Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Necroptosis hsa04217
Cell Process Cell ferroptosis
Cell necroptosis
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
In Vivo Model
Male Sprague-Dawley (SD) rats (250-270 g) were purchased from the Laboratory Animal Center, Xiangya, School of Medicine, Central South University, China. A left thoracotomy was carried out in the fourth intercostal space and the heart was exposed via opening the pericardium. Blockage of the left coronary artery was conducted via clamping the snare against the heart surface. Reperfusion was performed by release of the snare. 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 The combination of ponatinib with deferoxamine reduces myocardial ischemia/reperfusion (I/R) injury via simultaneous inhibition of necroptosis and ferroptosis.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [34]
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Drug Indolylmaleimide derivative IM-93 Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
NETosis
Response regulation The indolylmaleimide (IM) derivativeIM-17shows inhibitory activity against oxidative-stress-induced necrotic cell death and cardioprotective activity in rat ischemia-reperfusion injury models. IM-93 inhibited ferroptosis and NETosis, but not necroptosis or pyroptosis.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [35]
Responsed Disease Cold Ischemia/Reperfusion Injury [ICD-11: DB98]
Responsed Drug Liproxstatin-1 Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model BEAS-2B cells Normal Homo sapiens CVCL_0168
In Vivo Model
All animal procedures were approved and performed according to the protocols established by the Institutional Animal Care and Use Committee of Nanjing Medical University (approval number: 2022; June 2022). Male inbred C57BL/6J mice were purchased from Changzhou Cavans Animal Experiment Co., Ltd (Changzhou, China). Animals were housed in a temperature-controlled (22 ± 2 ) and humidity-controlled room with free access to both fresh water and standard laboratory food in Wuxi Peoples Hospital Animal Experiment Center. Ten- to 12-wk-old animals weighing 25 to 28 g were used for LTx.

    Click to Show/Hide
Response regulation Using liproxstatin-1 (Lip-1) to inhibit ferroptosis during CI could ameliorate LTx- cold ischemia-reperfusion injury, suggesting that Lip-1 administration might be proposed as a new strategy for organ preservation.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [36]
Responsed Disease Renal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator Cold-inducible RNA-binding protein (CIRBP) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model HK-2 cells Normal Homo sapiens CVCL_0302
In Vivo Model
Male C57BL/6 mice weighing 20-25 g were subjected to IR as described previously. Renal pedicles were accessed through a midline abdominal incision and clamped for 30 min. Mice body temperature was maintained at 32 throughout the procedure using a heatpad. Clamps were removed and the abdomen was closed after confirmation that blood flow had returned to the kidneys. Reperfusion was allowed to continue for 24 hours.

    Click to Show/Hide
Response regulation CIRBP promotes ferroptosis by interacting with ELAVL1 and activating ferritinophagy during renal ischaemia-reperfusion (IR) injury.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [37]
Responsed Disease Renal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator Fatty acyl-CoA reductase 1 (FAR1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HT-1080 cells Fibrosarcoma Homo sapiens CVCL_0317
786-O cells Renal cell carcinoma Homo sapiens CVCL_1051
MDA-MB-231 cells Breast adenocarcinoma Homo sapiens CVCL_0062
U-87MG cells Glioblastoma Homo sapiens CVCL_GP63
A2780 cells Ovarian endometrioid adenocarcinoma Homo sapiens CVCL_0134
HO8910 cells Endocervical adenocarcinoma Homo sapiens CVCL_6868
Hep-G2 cells Hepatoblastoma Homo sapiens CVCL_0027
Hep 3B2.1-7 cells Hepatocellular carcinoma Homo sapiens CVCL_0326
HuH-6 cells Hepatoblastoma Homo sapiens CVCL_4381
Huh-7 cells Hepatocellular carcinoma Homo sapiens CVCL_0336
Li-7 cells Adult hepatocellular carcinoma Homo sapiens CVCL_3840
In Vivo Model
8-week old female mice were purchased from Charles River Laboratories and prepared for the establishment of kidney ischemia/reperfusion. The mice were randomly distributed and anaesthetized with subcutaneous injection of sodium pentobarbital (30 mg/kg, Sigma). The heating pads were used to maintain the body temperature. Via the abdominal approach, the bilateral renal pedicles were clamped for 30 min by using a vascular clamp. Then the clamp was removed and the abdominal cavity was closed using sutures. For the group of ferrostatin-1 treatment, mice were injected intraperitoneally 200 ul PBS containing 5 mg/kg ferrostatin-1 30 min before ischemia.

    Click to Show/Hide
Response regulation FAR1 expression is positively correlated with the process of ferroptosis in renal ischemia reperfusion injury (IRI) and tumors. TMEM189, a newly identified gene which introduces vinyl-ether double bond into alkyl-ether lipids to generate plasmalogens abrogates FAR1-alkyl-ether lipids axis induced ferroptosis.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [38]
Responsed Disease Myocardial ischemia/reperfusion [ICD-11: DB98]
Responsed Regulator rno-miR-199a-5p (miRNA) Driver
Pathway Response Ferroptosis hsa04216
PI3K-Akt signaling pathway hsa04151
Cell Process Cell ferroptosis
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
Response regulation miR-199a-5 promotes ferroptosis-induced cardiomyocyte death via the inhibition of AKT/eNOS signaling pathway, thereby contributing to OGD/R injury, suggesting its potential as a target for the development of myocardial ischemia/reperfusion (I/R) injury and focusing on inhibition of miR-199a-5 may be beneficial for attenuating myocardial I/R injury.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [39]
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator ELAV-like protein 1 (ELAVL1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model hCMs (Human cardiomyocytes)
In Vivo Model
Healthy male C57BL/6J mice (8 weeks) were purchased from Shanghai SLAC laboratory Animal Co., Ltd. (Shanghai, China) and kept in the standard animal facility. To induce myocardial I/R injury, mice were anesthetized by ketamine and xylazine (250 and 10 mg/kg, respectively) first and maintained on 3% isoflurane. An abdominal incision was made around the fourth intercostal space and the left anterior descending coronary artery (LAD) was exposed. LAD was ligated with the silk suture for 1 h followed by 4 h of perfusion. During the perfusion, the incision was closed. For the sham group, same surgery procedures were performed without any ligation. For inhibition of ELAVL1 or overexpression of Beclin-1 in mice, lentivirus vectors (1 x 108 titers) obtained from GeneChem (Shanghai, China) were used for left ventricular cavity injection. After 7 days of lentivirus infection, mice were subjected to myocardial I/R surgery.

    Click to Show/Hide
Response regulation FOXC1 transcriptionally activated ELAVL1 may promote ferroptosis during myocardial ischemia and reperfusion (I/R) injury by modulating autophagy, leading to myocardial injury. Inhibition of ELAVL1-mediated autophagic ferroptosis would be a new viewpoint in the treatment of myocardial I/R injury.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [40]
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Indoleamine 2,3-dioxygenase 1 (IDO1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
In Vitro Model mRPTECs (Mouse renal proximal tubular epithelial cells)
Response regulation In RPTECs, both anoxia and reoxygenation upregulated IDO, which in turn induced GCN2K mediated apoptosis and AhR mediated ferroptosis. Since both phases of IR injury share IDO upregulation as a common point, its inhibition may prove a useful therapeutic strategy for preventing or attenuating ischemia reperfusion injury.
Transferrin receptor protein 1 (TFRC)
In total 4 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Cyanidin-3-glucoside Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell proliferation
Cell autophagy
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
In Vivo Model
Adult male Sprague Dawley (SD) rats weighing 260-280 g were purchased from Qinglongshan Animal Farm (Nanjing, China). After a week of adaptation, the rats were randomly assigned into five groups (n = 8): (1) sham group, rats receiving saline gavage and sham surgery were used as control group; (2) I/R model group, rats receiving saline gavage and left anterior descending (LAD) ligation surgery were used as the model group; (3) C3G-10 group, I/R model plus intraperitoneal injection of 10 mg/kg C3G; (4) C3G-20 group, I/R model plus intraperitoneal injection of 20 mg/kg C3G; and (5) DIL group, I/R model plus oral administration of 20 mg/kg diltiazem. C3G and DIL were dissolved in DMSO and then diluted with saline so that the DMSO concentration was less than 0.1% (v/v).

    Click to Show/Hide
Response regulation The administration of Cyanidin-3-glucoside (C3G) reduced the infarction area, mitigated pathological alterations, inhibited ST segment elevation, and attenuated oxidative stress and ferroptosis-related protein expression. C3G also suppressed the expressions of USP19, Beclin1, NCOA4, and LC3II/LC3I. In addition, treatment with C3G relieved oxidative stress, downregulated LC3II/LC3I, reduced autophagosome number, downregulated TfR1 expression, and upregulated the expressions of FTH1 and GPX4 in OGD/R-induced H9c2 cells. Taken together, C3G could be a potential agent to protect myocardium from myocardial ischemia-reperfusion (IR) injury.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [6]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Hepatic ischemia-reperfusion injury [ICD-11: DB98]
Responsed Regulator rno-miR-29a-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model rBMMSCs (Rat bone marrow mesenchymal stem cells)
IAR 20 cells Normal Homo sapiens CVCL_5296
In Vivo Model
Clean-grade male Sprague-Dawley (SD) rats were purchased from China Food and Drug Administration (Beijing, China). SD rats were fed a high-fat diet (Composition: 15% triglyceride, 15% sucrose, 10% egg yolk powder, 1% cholesterol, 0.2% bile salt, 58.8% basic feed) for 20 weeks. Hematoxylin and eosin (HE) and oil red O staining showed that the area of mixed macrovesicular steatosis was more than 60% under the microscope, indicating that a model of severe steatotic liver was established successfully. A 70% liver thermal ischemia model was established, continuously blocked for 80 min, and then, the ischemic liver was obtained 24 h after reperfusion.

    Click to Show/Hide
Response regulation miR-29a-3p, which targets IREB2, is abundant in HO-1/BMMSC-exosomes and could decrease the IREB2 protein level. The reduced IREB2 level led to an increase in the level of FTH1 and decreased the level of TFR1 through posttranscriptional regulation, which ultimately reduced the level of intracellular Fe2+ and the production of lipid ROS and inhibited the occurrence of ferroptosis in SHP-HR. In conclusion, ferroptosis plays an important role in HO-1/BMMSC-mediated alleviation of steatotic hepatic ischemia-reperfusion injury.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [7]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Ubiquitin carboxyl-terminal hydrolase 7 (USP7) Driver
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.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [7]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Cellular tumor antigen p53 (TP53) Driver
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.
Prostaglandin G/H synthase 2 (PTGS2)
In total 3 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Marker
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Baicalein Investigative
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
In this study, 30 male Sprague Dawley rats (325-375 g) anesthetized using pentobarbital (1.5 g/kg, i.p.) were used for heart infarct studies,Western blot analysis, and qPCR. The isolated hearts were perfused in a Langendorff system. A water-filled latex balloon was inserted into the left ventricle cavity via mitral valve and linked to a physiological pressure transducer (AD Instruments, MLT884) for continuous monitoring of left ventricular systolic pressure (LVSP) and end diastolic pressure (LVEDP). Left ventricular developed pressure (LVDP) was calculated as the difference between LVSP and LVEDP (LVDP = LVSP-LVEDP). Measurements were recorded using PowerLab system and Chart 8 software (ADInstrument, Bella Vista, New South Wales, Australia). The hearts were stable for 30 min, and then subjected to 45 min of global ischemia by halting perfusion, followed by 1 h of reperfusion with Krebs-Henseleit (KH) bicarbonate buffer gassed with 95% O2, 5% CO2 at 37 (pH 7.4). The infarcted myocardium was measured using triphenyltetrazolium chloride(TTC, 25 mg/mL) staining. The KH buffer containing 118 mM NaCl, 4.8 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 25 mM NaHCO 31.3 mM CaCl2, and 11 mM glucose was filtered through a 0.22 uM pore before use.

    Click to Show/Hide
Response regulation Baicalein and luteolin protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both baicalein and luteolin decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. Baicalein and luteolin reduced the ischemia/reperfusion-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Target for Ferroptosis Marker
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Histochrome Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model mCMs (Mouse cardiomyocytes)
In Vivo Model
Male Fischer 344 rats (8 weeks old and 160 to 180 g; KOATECH, Pyeongtaek-si, Korea) were anesthetized by inhalation with 2% isoflurane and intubated using an 18-gauge intravenous catheter. The rats were mechanically ventilated with medical-grade oxygen. Surgery was performed on a 37 heating pad to prevent the body from getting cold. A left thoracotomy was performed after the chest was shaved to prevent contamination during surgery.

    Click to Show/Hide
Response regulation Histochrome treatment significantly increased GPx4 and free GSH levels, but decreased Cox-2 level. HC treatment significantly decreased intracellular and mitochondrial ROS levels by upregulating the expression of Nrf2 and antioxidant genes. The substantial cardioprotective effects of HC against myocardia I/R injury by reducing ferroptosis-associated myocardial injury.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Marker
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Luteolin Investigative
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
In this study, 30 male Sprague Dawley rats (325-375 g) anesthetized using pentobarbital (1.5 g/kg, i.p.) were used for heart infarct studies,Western blot analysis, and qPCR. The isolated hearts were perfused in a Langendorff system. A water-filled latex balloon was inserted into the left ventricle cavity via mitral valve and linked to a physiological pressure transducer (AD Instruments, MLT884) for continuous monitoring of left ventricular systolic pressure (LVSP) and end diastolic pressure (LVEDP). Left ventricular developed pressure (LVDP) was calculated as the difference between LVSP and LVEDP (LVDP = LVSP-LVEDP). Measurements were recorded using PowerLab system and Chart 8 software (ADInstrument, Bella Vista, New South Wales, Australia). The hearts were stable for 30 min, and then subjected to 45 min of global ischemia by halting perfusion, followed by 1 h of reperfusion with Krebs-Henseleit (KH) bicarbonate buffer gassed with 95% O2, 5% CO2 at 37 (pH 7.4). The infarcted myocardium was measured using triphenyltetrazolium chloride(TTC, 25 mg/mL) staining. The KH buffer containing 118 mM NaCl, 4.8 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 25 mM NaHCO 31.3 mM CaCl2, and 11 mM glucose was filtered through a 0.22 uM pore before use.

    Click to Show/Hide
Response regulation Baicalein and luteolin protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both baicalein and luteolin decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. Baicalein and luteolin reduced the ischemia/reperfusion-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA.
Nuclear receptor coactivator 4 (NCOA4)
In total 3 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Cyanidin-3-glucoside Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell proliferation
Cell autophagy
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
In Vivo Model
Adult male Sprague Dawley (SD) rats weighing 260-280 g were purchased from Qinglongshan Animal Farm (Nanjing, China). After a week of adaptation, the rats were randomly assigned into five groups (n = 8): (1) sham group, rats receiving saline gavage and sham surgery were used as control group; (2) I/R model group, rats receiving saline gavage and left anterior descending (LAD) ligation surgery were used as the model group; (3) C3G-10 group, I/R model plus intraperitoneal injection of 10 mg/kg C3G; (4) C3G-20 group, I/R model plus intraperitoneal injection of 20 mg/kg C3G; and (5) DIL group, I/R model plus oral administration of 20 mg/kg diltiazem. C3G and DIL were dissolved in DMSO and then diluted with saline so that the DMSO concentration was less than 0.1% (v/v).

    Click to Show/Hide
Response regulation The administration of Cyanidin-3-glucoside (C3G) reduced the infarction area, mitigated pathological alterations, inhibited ST segment elevation, and attenuated oxidative stress and ferroptosis-related protein expression. C3G also suppressed the expressions of USP19, Beclin1, NCOA4, and LC3II/LC3I. In addition, treatment with C3G relieved oxidative stress, downregulated LC3II/LC3I, reduced autophagosome number, downregulated TfR1 expression, and upregulated the expressions of FTH1 and GPX4 in OGD/R-induced H9c2 cells. Taken together, C3G could be a potential agent to protect myocardium from myocardial ischemia-reperfusion (IR) injury.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [17]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug DNA (cytosine-5)-methyltransferase 1 Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
In Vivo Model
Fifty specific pathogen-free male SpragueDawley rats (weighing 210-240 g) were purchased from Beijing Huakang Biotechnology Co., Ltd (Beijing, China). The DS model was established by injecting 1% streptozotocin into the tail vein at 60 mg/kg dose. After 3 days, if the fasting blood glucose level was higher than 16.7 mmol/L, the DS model was successfully built. The NS and the I/R group were given 0.9% sodium chloride injection. Thereafter, the general conditions for normal and DM rats are showed in Table Table2.2. After 8 weeks, all the rats were intraperitoneally injected with 1.5% sodium pentobarbital at a dose of 0.005 mL/g. They were given electrocardiogram (ECG) monitoring management.

    Click to Show/Hide
Response regulation Inhibition of DNA (cytosine-5)-methyltransferase 1 (DNMT-1) could reduce ferroptosis during diabetes myocardial ischemia/reperfusion injury and the NCOA4-mediated ferritinophagy may participate in the process.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [18]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Nobiletin Investigative
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
Male Sprague-Dawley (SD) rats (4 weeks old, 90-110 g) were obtained from the Vital River Biological company. Rats were kept in a specific-pathogen-free (SPF) environment, with access to food and tap water at an ambient temperature of 20-22 . All institutional and national guidelines for the care and use of laboratory animals were followed. The protocols were reviewed and approved by the Institution of Animal Care and Use Committee of Renmin Hospital of Wuhan University (IACUC, license no. 20200303).

    Click to Show/Hide
Response regulation Both ferrostain-1 and nobiletin decreased the expression of ferroptosis-related proteins including Acyl-CoA synthetase long chain family member 4 (ACSL4) and nuclear receptor coactivator 4 (NCOA4) but not glutathione peroxidase 4 (GPX4) in rats with mature T2DM and cells with HFHG and H/R injury. Nobiletin has therapeutic potential for alleviating myocardial ischemia-reperfusion injury associated with ACSL4- and NCOA4-related ferroptosis.
Natural resistance-associated macrophage protein 2 (SLC11A2)
In total 2 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Target for Ferroptosis Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Metalloreductase STEAP3 (STEAP3) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model rBMMSCs (Rat bone marrow mesenchymal stem cells)
IAR 20 cells Normal Homo sapiens CVCL_5296
Response regulation MiR-124-3p in HM-exos downregulates Steap3 expression to inhibit ferroptosis, thereby attenuating graft ischemia reperfusion injury. And HUCB-MSCs-exos inhibited the expression of DMT1 by delivering miR-23a-3p, which suppressed cardiomyocyte ferroptosis after myocardial infarction.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Target for Ferroptosis Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator hsa-miR-124-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model rBMMSCs (Rat bone marrow mesenchymal stem cells)
IAR 20 cells Normal Homo sapiens CVCL_5296
Response regulation MiR-124-3p in HM-exos downregulates Steap3 expression to inhibit ferroptosis, thereby attenuating graft ischemia reperfusion injury. And HUCB-MSCs-exos inhibited the expression of DMT1 by delivering miR-23a-3p, which suppressed cardiomyocyte ferroptosis after myocardial infarction.
NADPH oxidase 4 (NOX4)
In total 2 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [25]
Target for Ferroptosis Driver
Responsed Disease Renal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator Lysine-specific histone demethylase 1A (KDM1A) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HK-2 cells Normal Homo sapiens CVCL_0302
In Vivo Model
Adult male C57BL6 (C57) mice (8-12 weeks, 20-25 g) were purchased from the Animal Experiment Center of Wuhan University. All 64 mice were randomly divided into various groups by different treatments (n = 8). In sham group, after the right kidney excised, the left renal pedicles were without any treatment. In IRI group, the pedicle of the left kidney was clamped for 30 min followed by various reperfusion periods (6, 12, 24 h). To study the effects of LSD1, TCP (MedChemExpress) was injected intraperitoneally at different doses (2.5, 5, 10 mg/kg) before IRI model establishment, once a day for 1 week. TCP powder was dissolved in dimethyl sulfoxide (DMSO). In the vehicle control group, equal amount of DMSO was injected intraperitoneally.

    Click to Show/Hide
Response regulation LSD1 (KDM1A) inhibition blocked ferroptosis and oxidative stress caused by renal IRI through the TLR4/NOX4 pathway, indicating that LSD1 could be a potential therapeutic target for renal ischaemia reperfusion injury (IRI).
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [25]
Target for Ferroptosis Driver
Responsed Disease Renal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator Toll-like receptor 4 (TLR4) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HK-2 cells Normal Homo sapiens CVCL_0302
In Vivo Model
Adult male C57BL6 (C57) mice (8-12 weeks, 20-25 g) were purchased from the Animal Experiment Center of Wuhan University. All 64 mice were randomly divided into various groups by different treatments (n = 8). In sham group, after the right kidney excised, the left renal pedicles were without any treatment. In IRI group, the pedicle of the left kidney was clamped for 30 min followed by various reperfusion periods (6, 12, 24 h). To study the effects of LSD1, TCP (MedChemExpress) was injected intraperitoneally at different doses (2.5, 5, 10 mg/kg) before IRI model establishment, once a day for 1 week. TCP powder was dissolved in dimethyl sulfoxide (DMSO). In the vehicle control group, equal amount of DMSO was injected intraperitoneally.

    Click to Show/Hide
Response regulation LSD1 (KDM1A) inhibition blocked ferroptosis and oxidative stress caused by renal IRI through the TLR4/NOX4 pathway, indicating that LSD1 could be a potential therapeutic target for renal ischaemia reperfusion injury (IRI).
Long-chain-fatty-acid--CoA ligase 4 (ACSL4)
In total 14 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Gossypol acetic acid Investigative
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
A total of 55 adult male Sprague-Dawley rat (350-450 g) were anesthetized with urethane (1.5 g/kg, i.p.), then the hearts were perfused in a Langendorff system. After 30 min of stabilization, hearts were subjected to 30 min of global no-flow ischemia by stopping the perfusion. Reperfusion was followed with Krebs Henseleit (KH) buffer and GAA together for 2 h. A thermoregulated chamber kept the heart at 37 throughout the experiment. Control hearts were not subjected to I/R. The heart slices were sectioned at a thickness of 2 mm and stained with triphenyltetrazolium chloride (25 mg/100 mL) for 10 min and then fixed with 4% formaldehyde solution for 48 h to enhance color contrast.

    Click to Show/Hide
Response regulation Gossypol acetic acid significantly attenuated myocardial infarct size, reduced lipid peroxidation, decreased the mRNA levels of the ferroptosis markers Ptgs2 and Acsl4, decreased the protein levels of ACSL4 and NRF2, and increased the protein levels of GPX4 in I/R-induced ex vivo rat hearts. Thus, GAA may play a cytoprotectant role in ferroptosis-induced cardiomyocyte death and myocardial ischemia/reperfusion-induced ferroptotic cell death.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [26]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Baicalin Terminated
Pathway Response Ferroptosis hsa04216
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell autophagy
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
In Vivo Model
Male Sprague-Dawley rats, 260-280 g, were provided by Beijing Vital River Laboratory Animal Technology CO., Ltd. (Beijing, China). Rats were randomly divided into five groups (n = 15 per group): control (sham operation + saline), I/R (I/R + saline), baicalin 100 mg/kg (BA-100, I/R + baicalin 100 mg/kg), baicalin 200 mg/kg (BA-200, I/R + baicalin 200 mg/kg), and diltiazem 20 mg/kg (DI-20, I/R + diltiazem 20 mg/kg). Drugs were given by oral gavage once daily (8 a.m.) for 6 days. At day 6, myocardial ischemia was induced 1 h after drug was administered.

    Click to Show/Hide
Response regulation Baicalin prevents against myocardial ischemia/reperfusion injury via suppressing ACSL4-controlled ferroptosis. In addition, enhanced lipid peroxidation and significant iron accumulation along with activated transferrin receptor protein 1 (TfR1) signal and nuclear receptor coactivator 4 (NCOA4)-medicated ferritinophagy were observed in in vivo and in vitro models, which were reversed by baicalin treatment.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Baicalein Investigative
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
In this study, 30 male Sprague Dawley rats (325-375 g) anesthetized using pentobarbital (1.5 g/kg, i.p.) were used for heart infarct studies,Western blot analysis, and qPCR. The isolated hearts were perfused in a Langendorff system. A water-filled latex balloon was inserted into the left ventricle cavity via mitral valve and linked to a physiological pressure transducer (AD Instruments, MLT884) for continuous monitoring of left ventricular systolic pressure (LVSP) and end diastolic pressure (LVEDP). Left ventricular developed pressure (LVDP) was calculated as the difference between LVSP and LVEDP (LVDP = LVSP-LVEDP). Measurements were recorded using PowerLab system and Chart 8 software (ADInstrument, Bella Vista, New South Wales, Australia). The hearts were stable for 30 min, and then subjected to 45 min of global ischemia by halting perfusion, followed by 1 h of reperfusion with Krebs-Henseleit (KH) bicarbonate buffer gassed with 95% O2, 5% CO2 at 37 (pH 7.4). The infarcted myocardium was measured using triphenyltetrazolium chloride(TTC, 25 mg/mL) staining. The KH buffer containing 118 mM NaCl, 4.8 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 25 mM NaHCO 31.3 mM CaCl2, and 11 mM glucose was filtered through a 0.22 uM pore before use.

    Click to Show/Hide
Response regulation Baicalein and luteolin protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both baicalein and luteolin decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. Baicalein and luteolin reduced the ischemia/reperfusion-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Target for Ferroptosis Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Drug Curcumin Investigative
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model hLCs (Liver cells)
rPTs (Rat pancreas tissues)
rHTs (Rat hippocampal tissues)
In Vivo Model
Forty female albino Wistar rats weighing 180-220 g were used in the study. Eight rats in each group were randomly assigned to five different groups: Group I (Sham); Group II (IR); Group III (IR + DMSO); Group IV (IR + Curcumin 100 mg/kg); and Group V (IR + 2 ug/kg LoxBlock-1) were determined. The animals were maintained at a temperature of 21 ± 2 and regulated humidity conditions (50 ± 5%) with a twelve-hour light/dark cycle. Throughout the experiment, the animals were fed standard commercial rat pellets and given tap water. All surgical and anesthesia procedures were performed understerile conditions. In addition, in a case of abnormal symptoms, the animals would be removed from the group and sacrificed under deep anesthesia.

    Click to Show/Hide
Response regulation Curcumin attenuates liver, pancreas and cardiac ferroptosis, oxidative stress and injury in ischemia/reperfusion-damaged rats by facilitating ACSL/GPx4 signaling.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Luteolin Investigative
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
In this study, 30 male Sprague Dawley rats (325-375 g) anesthetized using pentobarbital (1.5 g/kg, i.p.) were used for heart infarct studies,Western blot analysis, and qPCR. The isolated hearts were perfused in a Langendorff system. A water-filled latex balloon was inserted into the left ventricle cavity via mitral valve and linked to a physiological pressure transducer (AD Instruments, MLT884) for continuous monitoring of left ventricular systolic pressure (LVSP) and end diastolic pressure (LVEDP). Left ventricular developed pressure (LVDP) was calculated as the difference between LVSP and LVEDP (LVDP = LVSP-LVEDP). Measurements were recorded using PowerLab system and Chart 8 software (ADInstrument, Bella Vista, New South Wales, Australia). The hearts were stable for 30 min, and then subjected to 45 min of global ischemia by halting perfusion, followed by 1 h of reperfusion with Krebs-Henseleit (KH) bicarbonate buffer gassed with 95% O2, 5% CO2 at 37 (pH 7.4). The infarcted myocardium was measured using triphenyltetrazolium chloride(TTC, 25 mg/mL) staining. The KH buffer containing 118 mM NaCl, 4.8 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 25 mM NaHCO 31.3 mM CaCl2, and 11 mM glucose was filtered through a 0.22 uM pore before use.

    Click to Show/Hide
Response regulation Baicalein and luteolin protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both baicalein and luteolin decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. baicalein and luteolin reduced the ischemia/reperfusion-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [18]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Nobiletin Investigative
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
Male Sprague-Dawley (SD) rats (4 weeks old, 90-110 g) were obtained from the Vital River Biological company. Rats were kept in a specific-pathogen-free (SPF) environment, with access to food and tap water at an ambient temperature of 20-22 . All institutional and national guidelines for the care and use of laboratory animals were followed. The protocols were reviewed and approved by the Institution of Animal Care and Use Committee of Renmin Hospital of Wuhan University (IACUC, license no. 20200303).

    Click to Show/Hide
Response regulation Both ferrostain-1 and nobiletin decreased the expression of ferroptosis-related proteins including Acyl-CoA synthetase long chain family member 4 (ACSL4) and nuclear receptor coactivator 4 (NCOA4) but not glutathione peroxidase 4 (GPX4) in rats with mature T2DM and cells with HFHG and H/R injury. Nobiletin has therapeutic potential for alleviating myocardial ischemia-reperfusion injury associated with ACSL4- and NCOA4-related ferroptosis.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [27]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Xanthohumol 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
Rats were anesthetized with urethane (1.5 g/kg, i.p.), then hearts were excised and arrested in Krebs Henseleit (KH) buffer as previously described. Following 30 min equilibration, ischemia was induced by halting perfusion for 45 min. Reperfusion was followed with KH buffer and XN (5 or 10 uM) together for 60 min. Control hearts were not subjected to I/R.

    Click to Show/Hide
Response regulation Xanthohumol can prevent ferroptosis during cardiac ischemia-reperfusion by reducing the expression of Acsl4 and Ptgs2 mRNA, reducing the expression of ACSL4 and NRF2 protein, and modulating the expression of GPX4 protein.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [28]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion [ICD-11: DB98]
Responsed Regulator LncAABR07025387.1 (IncRNA) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
HEK-293T cells Normal Homo sapiens CVCL_0063
In Vivo Model
Adult male Sprague Dawley rats (3 months old, 200 g) purchased from the Animal Center of Nanjing University were housed at a controlled temperature of 18-22 and humidity of 50-70% under a 12-h light/dark cycle with free access to food and water. Before the operation, the rats were fasted overnight with free access to water. At the beginning of the operation, the rats were intraperitoneally injected with chloral hydrate (0.5 ml/100 g) for sedation) and 5% additional first dose for unsatisfactory sedation and made to inhale isoflurane for anesthesia. After incising the skin, the muscle was carefully separated layer by layer, and the trachea was exposed and fixed locally with a self-made pull hook. The trachea was then cut with a 10 ml syringe needle. The anesthesia mask was then replaced with the tracheal intubation. The ventilator was connected to the anesthesia machine for isopentane inhalation (2%). The proximal left anterior descending artery (LAD) was ligated using a 6.0 Prolene suture to induce myocardial ischemia. A small semi-cylindrical plastic hose was inserted to the LAD to facilitate the opening of the knot during reperfusion and to reduce the mechanical damage to the heart tissue and blood vessels. About 5 min later, the chest cavity was temporarily closed with a non-damaging hemostatic clip. After 30 min of ischemia, the noninjury hemostatic clip was loosened, the ligation and the protective tube were removed for reperfusion, and the chest was closed. The rats in the sham group underwent the same operation without being subjected to MI/R.

    Click to Show/Hide
Response regulation LncAABR07025387.1 acts as a competing endogenous RNA during myocardial ischemia/reperfusion injury. Mechanistically, lncAABR07025387.1 negatively regulates miR-205 expression and subsequently upregulates ACSL4-mediated ferroptosis.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator MIR9-3HG (IncRNA) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HL-1 cells Normal Mus musculus CVCL_0303
hBMSCs (Bone marrow stromal cells)
In Vivo Model
A total of 96 C57BL/6 male mice (20-25 g) aged 11-12 weeks were purchased from experimental animal center of experimental animal center of Guangdong Medical University. 96 mice were randomly divided into four groups (24 mice per group): Sham group (200 ul of PBS), Sham + BMSCs-Exo group (200 ul of BMSCs-Exo), I/R group (200 ul of PBS) and I/R + BMSCs-Exo group (200 ul of BMSCs-Exo). After 10 days of adaptive feeding, all mice were injected intraperitoneally with 0.4-0.5 mL/100 g 1%Pentobarbital Sodium. I/R and I/R + BMSCs-Exo group mice were subjected to cardiac I/R injury induced by ligation of the left anterior descending artery (LAD) for 30 min followed by 24 h reperfusion. Sham and Sham + BMSCs-Exo mice were sham treated and subjected to the same surgical procedures as I/R mice except that they did not receive ligation of the left anterior descending coronary artery.

    Click to Show/Hide
Response regulation Cellular ferroptosis is involved in the pathogenesis of Ischemia-Reperfusion Injury. BMSCs-Exo lncRNA Mir9-3hg can inhibit ferroptosis by modulating the Pum2/PRDX6 axis to exhibit cardioprotective effectsinvivoandinvitro. Silence of PRDX6 markedly decreased cell proliferation, GSH content and Gpx4 protein level, as well as prominently increased iron ion concentration and levels of ROS content and ACSL4 protein in H/R-treated HL-1 cells.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Pumilio homolog 2 (PUM2) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HL-1 cells Normal Mus musculus CVCL_0303
hBMSCs (Bone marrow stromal cells)
In Vivo Model
A total of 96 C57BL/6 male mice (20-25 g) aged 11-12 weeks were purchased from experimental animal center of experimental animal center of Guangdong Medical University. 96 mice were randomly divided into four groups (24 mice per group): Sham group (200 ul of PBS), Sham + BMSCs-Exo group (200 ul of BMSCs-Exo), I/R group (200 ul of PBS) and I/R + BMSCs-Exo group (200 ul of BMSCs-Exo). After 10 days of adaptive feeding, all mice were injected intraperitoneally with 0.4-0.5 mL/100 g 1%Pentobarbital Sodium. I/R and I/R + BMSCs-Exo group mice were subjected to cardiac I/R injury induced by ligation of the left anterior descending artery (LAD) for 30 min followed by 24 h reperfusion. Sham and Sham + BMSCs-Exo mice were sham treated and subjected to the same surgical procedures as I/R mice except that they did not receive ligation of the left anterior descending coronary artery.

    Click to Show/Hide
Response regulation Cellular ferroptosis is involved in the pathogenesis of Ischemia-Reperfusion Injury. BMSCs-Exo lncRNA Mir9-3hg can inhibit ferroptosis by modulating the Pum2/PRDX6 axis to exhibit cardioprotective effectsinvivoandinvitro. Silence of PRDX6 markedly decreased cell proliferation, GSH content and Gpx4 protein level, as well as prominently increased iron ion concentration and levels of ROS content and ACSL4 protein in H/R-treated HL-1 cells.
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Driver
Responsed Disease Ischemia-Reperfusion Injury [ICD-11: DB98]
Responsed Regulator Peroxiredoxin-6 (PRDX6) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HL-1 cells Normal Mus musculus CVCL_0303
hBMSCs (Bone marrow stromal cells)
In Vivo Model
A total of 96 C57BL/6 male mice (20-25 g) aged 11-12 weeks were purchased from experimental animal center of experimental animal center of Guangdong Medical University. 96 mice were randomly divided into four groups (24 mice per group): Sham group (200 ul of PBS), Sham + BMSCs-Exo group (200 ul of BMSCs-Exo), I/R group (200 ul of PBS) and I/R + BMSCs-Exo group (200 ul of BMSCs-Exo). After 10 days of adaptive feeding, all mice were injected intraperitoneally with 0.4-0.5 mL/100 g 1%Pentobarbital Sodium. I/R and I/R + BMSCs-Exo group mice were subjected to cardiac I/R injury induced by ligation of the left anterior descending artery (LAD) for 30 min followed by 24 h reperfusion. Sham and Sham + BMSCs-Exo mice were sham treated and subjected to the same surgical procedures as I/R mice except that they did not receive ligation of the left anterior descending coronary artery.

    Click to Show/Hide
Response regulation Cellular ferroptosis is involved in the pathogenesis of Ischemia-Reperfusion Injury. BMSCs-Exo lncRNA Mir9-3hg can inhibit ferroptosis by modulating the Pum2/ PRDX6 axis to exhibit cardioprotective effectsinvivoandinvitro. Silence of PRDX6 markedly decreased cell proliferation, GSH content and Gpx4 protein level, as well as prominently increased iron ion concentration and levels of ROS content and ACSL4 protein in H/R-treated HL-1 cells.
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target [29]
Target for Ferroptosis Driver
Responsed Disease Ischemic/reperfusion injury [ICD-11: DB98]
Responsed Regulator TUG1 (IncRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HK-2 cells Normal Homo sapiens CVCL_0302
In Vivo Model
Mouse renal I/R model was performed in male C57BL/6 mice (8-12 weeks old). Briefly, the mice were anesthetized with pentobarbital sodium by intraperitoneal injection and lay on the right side. Dorsal incisions of both left and right sides were made to expose kidneys. The right kidney artery was gently separated with cotton swabs and occluded with a microvascular clamp to induce renal ischemia for 45 min. The left renal pedicle clamping and ischemia were the same as right. After ischemia, the micro-aneurysm clips were removed to start the reperfusion. The wounds were sutured and resuscitated with warm sterile saline intraperitoneally. All operations were the same in the sham group except for clamping and ischemia.

    Click to Show/Hide
Response regulation Human urine-derived stem cells (USCs)-derived exosomes (USC-Exo) could improve kidney ischemia/reperfusion injury (IRI). Mechanistically, LncRNA TUG1 was carried by USC-Exo downregulation of ACSL4 expression in kidney cells by interacting with SRSF1, then inhibited ACSL4-mediated cell ferroptosis, and thus improved kidney injury in IRI-induced AKI.
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target [29]
Target for Ferroptosis Driver
Responsed Disease Ischemic/reperfusion injury [ICD-11: DB98]
Responsed Regulator Serine/arginine-rich splicing factor 1 (SRSF1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HK-2 cells Normal Homo sapiens CVCL_0302
In Vivo Model
Mouse renal I/R model was performed in male C57BL/6 mice (8-12 weeks old). Briefly, the mice were anesthetized with pentobarbital sodium by intraperitoneal injection and lay on the right side. Dorsal incisions of both left and right sides were made to expose kidneys. The right kidney artery was gently separated with cotton swabs and occluded with a microvascular clamp to induce renal ischemia for 45 min. The left renal pedicle clamping and ischemia were the same as right. After ischemia, the micro-aneurysm clips were removed to start the reperfusion. The wounds were sutured and resuscitated with warm sterile saline intraperitoneally. All operations were the same in the sham group except for clamping and ischemia.

    Click to Show/Hide
Response regulation Human urine-derived stem cells (USCs)-derived exosomes (USC-Exo) could improve kidney ischemia/reperfusion injury (IRI). Mechanistically, LncRNA TUG1 was carried by USC-Exo downregulation of ACSL4 expression in kidney cells by interacting with SRSF1, then inhibited ACSL4-mediated cell ferroptosis, and thus improved kidney injury in IRI-induced AKI.
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target [28]
Target for Ferroptosis Driver
Responsed Disease Myocardial ischemia/reperfusion [ICD-11: DB98]
Responsed Regulator hsa-miR-205-3p (miRNA) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model CHO-S/H9C2 cells Normal Cricetulus griseus CVCL_A0TS
HEK-293T cells Normal Homo sapiens CVCL_0063
In Vivo Model
Adult male Sprague Dawley rats (3 months old, 200 g) purchased from the Animal Center of Nanjing University were housed at a controlled temperature of 18-22 and humidity of 50-70% under a 12-h light/dark cycle with free access to food and water. Before the operation, the rats were fasted overnight with free access to water. At the beginning of the operation, the rats were intraperitoneally injected with chloral hydrate (0.5 ml/100 g) for sedation) and 5% additional first dose for unsatisfactory sedation and made to inhale isoflurane for anesthesia. After incising the skin, the muscle was carefully separated layer by layer, and the trachea was exposed and fixed locally with a self-made pull hook. The trachea was then cut with a 10 ml syringe needle. The anesthesia mask was then replaced with the tracheal intubation. The ventilator was connected to the anesthesia machine for isopentane inhalation (2%). The proximal left anterior descending artery (LAD) was ligated using a 6.0 Prolene suture to induce myocardial ischemia. A small semi-cylindrical plastic hose was inserted to the LAD to facilitate the opening of the knot during reperfusion and to reduce the mechanical damage to the heart tissue and blood vessels. About 5 min later, the chest cavity was temporarily closed with a non-damaging hemostatic clip. After 30 min of ischemia, the noninjury hemostatic clip was loosened, the ligation and the protective tube were removed for reperfusion, and the chest was closed. The rats in the sham group underwent the same operation without being subjected to MI/R.

    Click to Show/Hide
Response regulation LncAABR07025387.1 acts as a competing endogenous RNA during myocardial ischemia/reperfusion injury. Mechanistically, lncAABR07025387.1 negatively regulates miR-205 expression and subsequently upregulates ACSL4-mediated ferroptosis.
Iron-responsive element-binding protein 2 (IREB2)
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 Hepatic ischemia-reperfusion injury [ICD-11: DB98]
Responsed Regulator rno-miR-29a-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model rBMMSCs (Rat bone marrow mesenchymal stem cells)
IAR 20 cells Normal Homo sapiens CVCL_5296
In Vivo Model
Clean-grade male Sprague-Dawley (SD) rats were purchased from China Food and Drug Administration (Beijing, China). SD rats were fed a high-fat diet (Composition: 15% triglyceride, 15% sucrose, 10% egg yolk powder, 1% cholesterol, 0.2% bile salt, 58.8% basic feed) for 20 weeks. Hematoxylin and eosin (HE) and oil red O staining showed that the area of mixed macrovesicular steatosis was more than 60% under the microscope, indicating that a model of severe steatotic liver was established successfully. A 70% liver thermal ischemia model was established, continuously blocked for 80 min, and then, the ischemic liver was obtained 24 h after reperfusion.

    Click to Show/Hide
Response regulation miR-29a-3p, which targets IREB2, is abundant in HO-1/BMMSC-exosomes and could decrease the IREB2 protein level. The reduced IREB2 level led to an increase in the level of FTH1 and decreased the level of TFR1 through posttranscriptional regulation, which ultimately reduced the level of intracellular Fe2+ and the production of lipid ROS and inhibited the occurrence of ferroptosis in SHP-HR. In conclusion, ferroptosis plays an important role in HO-1/BMMSC-mediated alleviation of steatotic hepatic ischemia-reperfusion injury.
Heme oxygenase 1 (HMOX1)
In total 4 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [19]
Target for Ferroptosis Suppressor
Responsed Disease Hepatic ischemia-reperfusion injury [ICD-11: DB98]
Responsed Drug Dimethyl fumarate Approved
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model AML12 cells Normal Mus musculus CVCL_0140
In Vivo Model
The mice were randomly divided into four groups of six: sham + vehicle, sham + DMF, IR + vehicle, and IR + DMF. The mice were supplemented with DMF at a concentration of 100 mg/kg or DMSO by daily oral gavage for a week before surgery, as previously reported. As stated in a prior study, the partial warm liver IRI model was developed. Briefly, the sham group only had free hepatic portal blood vessels after laparotomy, and the blood flow was not obstructed. As for the hepatic IR group, the blood supply to the left and mid-hepatic lobes was blocked, resulting in 70% mouse liver IRI for 90 min. The mice were put on a heated blanket after surgery in order to maintain body temperature and monitor vital signs. Blood supply was restored for 6 h. Died mice were eliminated for testing prior to sample collection. The mice were euthanized after the sample were obtained. The same experimenter carried out all surgeries.

    Click to Show/Hide
Response regulation NRF2 knockdown notably decreased the expression of SLC7A11 and HO-1 and blocked the anti-ferroptosis effects of dimethyl fumarate (DMF). DMF inhibits ferroptosis by activating the NRF2/SLC7A11/HO-1 axis and exerts a protective effect against hepatic ischemia-reperfusion injury.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [3]
Target for Ferroptosis Suppressor
Responsed Disease Intestinal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Drug Apigenin Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HUVECs (Human umbilical vein endothelial cells)
In Vivo Model
Mice were exposed to 12/12 h of light/darkness and given free access to food and water. All C57BL/6J mice were fasted for 12 h and anesthetized by intraperitoneal injection of 1% sodium pentobarbital before modeling. Later, the superior mesenteric artery (SMA) was subjected to 45 min of global no-flow ischemia, followed by 90 min of reperfusion to induce IIRI. The successful model could be revealed by the microcirculation detector. In order to investigate the effects of APG, mice were randomly divided into different groups: Sham group, IIRI group, APG groups (2 mg/kg, 4 mg/kg and 8 mg/kg), ZnPPIX group (Protoporphyrin Zinc(), Dalian Meilun Biotech Co., Ltd., China, 10mg/kg), Selegiline group (10 mg/kg, Shandong Topscience Biotech Co., Ltd., China) and the ZnPPIX + Selegiline group (10 mg/kg ZnPPIX and 10 mg/kg Selegiline). As mentioned above, drug was intraperitoneal injected after a 10-min-ischemia.

    Click to Show/Hide
Response regulation MST analysis suggested that Apigenin could specifically bind to heme oxygenase 1 (HO-1) and monoamine oxidase b (MAO-B). Simultaneously, APG could attenuate ROS generation and Fe2+ accumulation, maintain mitochondria function thus inhibit ferroptosis and alleviate intestinal ischemia-reperfusion injury.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [21]
Target for Ferroptosis Suppressor
Responsed Disease Lung ischemia/reperfusion Injury [ICD-11: DB98]
Responsed Drug Iridin Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model MLE-12 cells Normal Mus musculus CVCL_3751
In Vivo Model
In vivo, the LIRI model was established as described earlier. All mice were anesthetized with pentobarbital administered intraperitoneally (50 mg/kg, Sigma-Aldrich, MO, USA). After endotracheal intubation, the mice were ventilated using a rodent ventilator (MiniVent, Harvard Apparatus, USA), with the title volume set to 7 ml/kg, the respiratory rate set to 120 times/min, and the inspiratory/expiratory ratio set to 1: 2. A noninvasive clamp was used to interrupt the left pulmonary hilum, causing lung ischemia. The clamp was released after 60 minutes of ischemia, and the left lung was reperfused for 120 minutes. Animals were euthanized via cervical dislocation at the end of the experiment. Following that, lung specimens and bronchoalveolar lavage fluid were harvested for analysis. All procedures except lung ischemia were performed on mice in the sham group.

    Click to Show/Hide
Response regulation As a result, irisin postconditioning may protect against lung I/R damage by suppressing ferroptosis via the Nrf2/HO-1 signaling axis.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [30]
Target for Ferroptosis Suppressor
Responsed Disease Renal ischemia/reperfusion injury [ICD-11: DB98]
Responsed Regulator Pannexin-1 (PANX1) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Autophagy hsa04140
Cell Process Cell ferroptosis
Cell apoptosis
Cell autophagy
In Vitro Model HK-2 cells Normal Homo sapiens CVCL_0302
In Vivo Model
C57BL/6 mice (male, 10-15 weeks old) were food-deprived for 12 h before the procedures and were anesthetized with intraperitoneal injection of 1% sodium pentobarbital solution (40 mg/kg). Using a midline abdominal incision, bilateral renal IRI was induced by clamping renal pedicles for 30 min. After removal of the clamp, the kidneys were inspected to confirm reperfusion.

    Click to Show/Hide
Response regulation Panx1 deletion induced the expression of a cytoprotective chaperone, heme oxygenase-1 (HO-1), and inhibited ferroptinophagy via the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway. In summary, Panx1 deletion protects against renal ischemia/reperfusion injury (IRI) by attenuating MAPK/ERK activation in a ferroptotic pathway.
Ferritin heavy chain (FTH1)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [6]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Hepatic ischemia-reperfusion injury [ICD-11: DB98]
Responsed Regulator rno-miR-29a-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model rBMMSCs (Rat bone marrow mesenchymal stem cells)
IAR 20 cells Normal Homo sapiens CVCL_5296
In Vivo Model
Clean-grade male Sprague-Dawley (SD) rats were purchased from China Food and Drug Administration (Beijing, China). SD rats were fed a high-fat diet (Composition: 15% triglyceride, 15% sucrose, 10% egg yolk powder, 1% cholesterol, 0.2% bile salt, 58.8% basic feed) for 20 weeks. Hematoxylin and eosin (HE) and oil red O staining showed that the area of mixed macrovesicular steatosis was more than 60% under the microscope, indicating that a model of severe steatotic liver was established successfully. A 70% liver thermal ischemia model was established, continuously blocked for 80 min, and then, the ischemic liver was obtained 24 h after reperfusion.

    Click to Show/Hide
Response regulation miR-29a-3p, which targets IREB2, is abundant in HO-1/BMMSC-exosomes and could decrease the IREB2 protein level. The reduced IREB2 level led to an increase in the level of FTH1 and decreased the level of TFR1 through posttranscriptional regulation, which ultimately reduced the level of intracellular Fe2+ and the production of lipid ROS and inhibited the occurrence of ferroptosis in SHP-HR. In conclusion, ferroptosis plays an important role in HO-1/BMMSC-mediated alleviation of steatotic hepatic ischemia-reperfusion injury.
References
Ref 1 The gut microbiota metabolite capsiate promotes Gpx4 expression by activating TRPV1 to inhibit intestinal ischemia reperfusion-induced ferroptosis. Gut Microbes. 2021 Jan-Dec;13(1):1-21. doi: 10.1080/19490976.2021.1902719.
Ref 2 Inhibition of MALT1 reduces ferroptosis in rat hearts following ischemia/reperfusion via enhancing the Nrf2/SLC7A11 pathway. Eur J Pharmacol. 2023 Jul 5;950:175774. doi: 10.1016/j.ejphar.2023.175774. Epub 2023 May 3.
Ref 3 Old targets, new strategy: Apigenin-7-O--d-(-6-p-coumaroyl)-glucopyranoside prevents endothelial ferroptosis and alleviates intestinal ischemia-reperfusion injury through HO-1 and MAO-B inhibition. Free Radic Biol Med. 2022 May 1;184:74-88. doi: 10.1016/j.freeradbiomed.2022.03.033. Epub 2022 Apr 7.
Ref 4 Lidocaine attenuates hypoxia/reoxygenationinduced inflammation, apoptosis and ferroptosis in lung epithelial cells by regulating the p38 MAPK pathway. Mol Med Rep. 2022 May;25(5):150. doi: 10.3892/mmr.2022.12666. Epub 2022 Mar 4.
Ref 5 The Protective Effect of Cyanidin-3-Glucoside on Myocardial Ischemia-Reperfusion Injury through Ferroptosis. Oxid Med Cell Longev. 2021 Feb 6;2021:8880141. doi: 10.1155/2021/8880141. eCollection 2021.
Ref 6 miR-29a-3p in Exosomes from Heme Oxygenase-1 Modified Bone Marrow Mesenchymal Stem Cells Alleviates Steatotic Liver Ischemia-Reperfusion Injury in Rats by Suppressing Ferroptosis via Iron Responsive Element Binding Protein 2. Oxid Med Cell Longev. 2022 Jun 9;2022:6520789. doi: 10.1155/2022/6520789. eCollection 2022.
Ref 7 Ubiquitin-specific protease 7 promotes ferroptosis via activation of the p53/TfR1 pathway in the rat hearts after ischemia/reperfusion. Free Radic Biol Med. 2021 Jan;162:339-352. doi: 10.1016/j.freeradbiomed.2020.10.307. Epub 2020 Nov 4.
Ref 8 Baicalein and luteolin inhibit ischemia/reperfusion-induced ferroptosis in rat cardiomyocytes. Int J Cardiol. 2023 Mar 15;375:74-86. doi: 10.1016/j.ijcard.2022.12.018. Epub 2022 Dec 10.
Ref 9 Histochrome Attenuates Myocardial Ischemia-Reperfusion Injury by Inhibiting Ferroptosis-Induced Cardiomyocyte Death. Antioxidants (Basel). 2021 Oct 15;10(10):1624. doi: 10.3390/antiox10101624.
Ref 10 Gossypol Acetic Acid Attenuates Cardiac Ischemia/Reperfusion Injury in Rats via an Antiferroptotic Mechanism. Biomolecules. 2021 Nov 10;11(11):1667. doi: 10.3390/biom11111667.
Ref 11 Hydroxysafflor yellow A and anhydrosafflor yellow B alleviate ferroptosis and parthanatos in PC12cells injured by OGD/R. Free Radic Biol Med. 2022 Feb 1;179:1-10. doi: 10.1016/j.freeradbiomed.2021.12.262. Epub 2021 Dec 16.
Ref 12 LoxBlock-1 or Curcumin attenuates liver, pancreas and cardiac ferroptosis, oxidative stress and injury in Ischemia/reperfusion-damaged rats by facilitating ACSL/GPx4 signaling. Tissue Cell. 2023 Jun;82:102114. doi: 10.1016/j.tice.2023.102114. Epub 2023 May 18.
Ref 13 miR-135b-3p Promotes Cardiomyocyte Ferroptosis by Targeting GPX4 and Aggravates Myocardial Ischemia/Reperfusion Injury. Front Cardiovasc Med. 2021 Aug 13;8:663832. doi: 10.3389/fcvm.2021.663832. eCollection 2021.
Ref 14 MiR-375-3p Promotes Cardiac Fibrosis by Regulating the Ferroptosis Mediated by GPX4. Comput Intell Neurosci. 2022 Apr 22;2022:9629158. doi: 10.1155/2022/9629158. eCollection 2022.
Ref 15 The BMSC-derived exosomal lncRNA Mir9-3hg suppresses cardiomyocyte ferroptosis in ischemia-reperfusion mice via the Pum2/PRDX6 axis. Nutr Metab Cardiovasc Dis. 2022 Feb;32(2):515-527. doi: 10.1016/j.numecd.2021.10.017. Epub 2021 Nov 3.
Ref 16 Downregulation of TNFAIP1 alleviates OGD/Rinduced neuronal damage by suppressing Nrf2/GPX4mediated ferroptosis. Exp Ther Med. 2022 Nov 23;25(1):25. doi: 10.3892/etm.2022.11724. eCollection 2023 Jan.
Ref 17 Inhibition of DNMT-1 alleviates ferroptosis through NCOA4 mediated ferritinophagy during diabetes myocardial ischemia/reperfusion injury. Cell Death Discov. 2021 Sep 29;7(1):267. doi: 10.1038/s41420-021-00656-0.
Ref 18 Nobiletin alleviates myocardial ischemia-reperfusion injury via ferroptosis in rats with type-2 diabetes mellitus. Biomed Pharmacother. 2023 Jul;163:114795. doi: 10.1016/j.biopha.2023.114795. Epub 2023 May 3.
Ref 19 Dimethyl fumarate protects against hepatic ischemia-reperfusion injury by alleviating ferroptosis via the NRF2/SLC7A11/HO-1 axis. Cell Cycle. 2023 Apr;22(7):818-828. doi: 10.1080/15384101.2022.2155016. Epub 2022 Dec 8.
Ref 20 Etomidate Attenuates the Ferroptosis in Myocardial Ischemia/Reperfusion Rat Model via Nrf2/HO-1 Pathway. Shock. 2021 Sep 1;56(3):440-449. doi: 10.1097/SHK.0000000000001751.
Ref 21 Postconditioning with Irisin Attenuates Lung Ischemia/Reperfusion Injury by Suppressing Ferroptosis via Induction of the Nrf2/HO-1 Signal Axis. Oxid Med Cell Longev. 2022 Mar 2;2022:9911167. doi: 10.1155/2022/9911167. eCollection 2022.
Ref 22 Naringenin alleviates myocardial ischemia/reperfusion injury by regulating the nuclear factor-erythroid factor 2-related factor 2 (Nrf2) /System xc-/ glutathione peroxidase 4 (GPX4) axis to inhibit ferroptosis. Bioengineered. 2021 Dec;12(2):10924-10934. doi: 10.1080/21655979.2021.1995994.
Ref 23 Salidroside postconditioning attenuates ferroptosis-mediated lung ischemia-reperfusion injury by activating the Nrf2/SLC7A11 signaling axis. Int Immunopharmacol. 2023 Feb;115:109731. doi: 10.1016/j.intimp.2023.109731. Epub 2023 Jan 20.
Ref 24 miR-124-3p delivered by exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells inhibits ferroptosis to attenuate ischemia-reperfusion injury in steatotic grafts. J Nanobiotechnology. 2022 Apr 22;20(1):196. doi: 10.1186/s12951-022-01407-8.
Ref 25 Lysine-specific demethylase 1 aggravated oxidative stress and ferroptosis induced by renal ischemia and reperfusion injury through activation of TLR4/NOX4 pathway in mice. J Cell Mol Med. 2022 Aug;26(15):4254-4267. doi: 10.1111/jcmm.17444. Epub 2022 Jun 30.
Ref 26 Baicalin Prevents Myocardial Ischemia/Reperfusion Injury Through Inhibiting ACSL4 Mediated Ferroptosis. Front Pharmacol. 2021 Apr 14;12:628988. doi: 10.3389/fphar.2021.628988. eCollection 2021.
Ref 27 Xanthohumol Protects the Rat Myocardium against Ischemia/Reperfusion Injury-Induced Ferroptosis. Oxid Med Cell Longev. 2022 Jan 17;2022:9523491. doi: 10.1155/2022/9523491. eCollection 2022.
Ref 28 LncAABR07025387.1 Enhances Myocardial Ischemia/Reperfusion Injury Via miR-205/ACSL4-Mediated Ferroptosis. Front Cell Dev Biol. 2022 Feb 2;10:672391. doi: 10.3389/fcell.2022.672391. eCollection 2022.
Ref 29 Exosomal lncRNA TUG1 derived from human urine-derived stem cells attenuates renal ischemia/reperfusion injury by interacting with SRSF1 to regulate ASCL4-mediated ferroptosis. Stem Cell Res Ther. 2022 Jul 15;13(1):297. doi: 10.1186/s13287-022-02986-x.
Ref 30 Pannexin 1 mediates ferroptosis that contributes to renal ischemia/reperfusion injury. J Biol Chem. 2019 Dec 13;294(50):19395-19404. doi: 10.1074/jbc.RA119.010949. Epub 2019 Nov 6.
Ref 31 Circulating small extracellular vesicle-encapsulated SEMA5A-IT1 attenuates myocardial ischemia-reperfusion injury after cardiac surgery with cardiopulmonary bypass. Cell Mol Biol Lett. 2022 Oct 25;27(1):95. doi: 10.1186/s11658-022-00395-9.
Ref 32 MicroRNA-27a Regulates Ferroptosis Through SLC7A11 to Aggravate Cerebral ischemia-reperfusion Injury. Neurochem Res. 2023 May;48(5):1370-1381. doi: 10.1007/s11064-022-03826-3. Epub 2022 Dec 2.
Ref 33 Combination of ponatinib with deferoxamine synergistically mitigates ischemic heart injury via simultaneous prevention of necroptosis and ferroptosis. Eur J Pharmacol. 2021 May 5;898:173999. doi: 10.1016/j.ejphar.2021.173999. Epub 2021 Mar 3.
Ref 34 Development of a Water-Soluble Indolylmaleimide Derivative IM-93 Showing Dual Inhibition of Ferroptosis and NETosis. ACS Med Chem Lett. 2019 Jul 30;10(9):1272-1278. doi: 10.1021/acsmedchemlett.9b00142. eCollection 2019 Sep 12.
Ref 35 Liproxstatin-1 Alleviates Lung Transplantation-induced Cold Ischemia-Reperfusion Injury by Inhibiting Ferroptosis. Transplantation. 2023 May 19. doi: 10.1097/TP.0000000000004638. Online ahead of print.
Ref 36 CIRBP promotes ferroptosis by interacting with ELAVL1 and activating ferritinophagy during renal ischaemia-reperfusion injury. J Cell Mol Med. 2021 Jun 10;25(13):6203-16. doi: 10.1111/jcmm.16567. Online ahead of print.
Ref 37 Peroxisome-driven ether-linked phospholipids biosynthesis is essential for ferroptosis. Cell Death Differ. 2021 Aug;28(8):2536-2551. doi: 10.1038/s41418-021-00769-0. Epub 2021 Mar 17.
Ref 38 MiR-199a-5p promotes ferroptosis-induced cardiomyocyte death responding to oxygen-glucose deprivation/reperfusion injury via inhibiting Akt/eNOS signaling pathway. Kaohsiung J Med Sci. 2022 Nov;38(11):1093-1102. doi: 10.1002/kjm2.12605. Epub 2022 Oct 18.
Ref 39 ELAVL1 is transcriptionally activated by FOXC1 and promotes ferroptosis in myocardial ischemia/reperfusion injury by regulating autophagy. Mol Med. 2021 Feb 10;27(1):14. doi: 10.1186/s10020-021-00271-w.
Ref 40 Role of indoleamine 2,3-dioxygenase in ischemia-reperfusion injury of renal tubular epithelial cells. Mol Med Rep. 2021 Jun;23(6):472. doi: 10.3892/mmr.2021.12111. Epub 2021 Apr 26.