Ferroptosis-centered Disease Response Information
General Information of the Disease (ID: DIS00006)
Name |
Glioblastoma
|
||||
---|---|---|---|---|---|
ICD |
ICD-11: 2A00
|
Full List of Target(s) of This Ferroptosis-centered Disease
Solute carrier family 40 member 1 (SLC40A1)
In total 4 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 ( MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 (MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [13] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Regulator | Nuclear factor NF-kappa-B p100 subunit (NFKB2) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
NF-kappa B signaling pathway | hsa04064 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Royan N9 cells | Normal | Mus musculus | CVCL_9455 | |
Royan N33 cells | Normal | Mus musculus | CVCL_9417 | ||
T98 cells | Glioblastoma | Homo sapiens | CVCL_B368 | ||
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | ||
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
U118 cells | Astrocytoma | Homo sapiens | CVCL_0633 | ||
In Vivo Model |
Four-to five-week-old female BALB/c nude mice were obtained from the Laboratory Animal Center, Southern Medical University. To study the role of IRP1 in TMZ resistance, the mice were randomly divided into four groups (n = 6 per group) (U87TR, U87TR + TMZ, U87TR-lvIRP1, U87TR-lvIRP1 + TMZ). To establish the GBM models, IRP1 overexpress or control U87TR cells (5 x 105 cells per mice in 3 uL PBS) transfected with luciferase lentivirus were injected into the mice brain under the guidance of a stereotactic instrument at coordinates relative to bregma: 2.0 mm posterior, 2.0 mm lateral, and 3.0 mm ventral.
Click to Show/Hide
|
||||
Response regulation | Amplifying IRP1 signals can reverse TMZ resistance and suppress tumor growthin vivovia inhibiting NFKB2 in the noncanonical NF-kB signaling pathway. In addition, NFKB2 affected TMZ sensitivity of glioblastoma by modulating the expression of LCN2 and FPN1. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [14] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Regulator | hsa-miR-147a (miRNA) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Glutathione metabolism | hsa00480 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 | |
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
hMGCs (Human normal brain astroglia cells) | |||||
SVG p12 cells | Normal | Homo sapiens | CVCL_3797 | ||
Response regulation | miR-147a targets SLC40A1 to induce ferroptosis in human glioblastoma in vitro. Mechanistically, miR-147a directly bound to the 3'-untranslated region of SLC40A1 and inhibited SLC40A1-mediated iron export, thereby facilitating iron overload, lipid peroxidation, and ferroptosis. | ||||
Phospholipid hydroperoxide glutathione peroxidase (GPX4)
In total 15 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [2] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Peoniflorin | Investigative | |||
Responsed Regulator | E3 ubiquitin-protein ligase NEDD4-like (NEDD4L) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
In Vivo Model |
U251 cells (6 x 106) were inoculated into the flanks of 4-to 5-week-old athymic nude mice (Shanghai Laboratory Animal Company, Shanghai, China) subcutaneously to generate a subcutaneous xenograft tumor model. After 2 weeks, the tumor model was successfully constructed, the mice were treated single and combined with 100 mg/kg RSL3 (2 times/week) and 1.0 g/kg/days PF. Tumor volumes were measured every 4 days to draw the growth curve. Mice were sacrificed 4 weeks after cell injection. Tumor xenografts were collected, photographed, and weighed and the tumor apoptosis was analyzed by Tunel staining.
Click to Show/Hide
|
||||
Response regulation | Paeoniflorin (PF) can function as an antitumor agent for glioma treatment by targeting NEDD4L-dependent STAT3 ubiquitination as well as by regulating the Nrf2/GPX4 signaling axis, which might trigger ferroptosis. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [2] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Peoniflorin | Investigative | |||
Responsed Regulator | Signal transducer and activator of transcription 3 (STAT3) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
In Vivo Model |
U251 cells (6 x 106) were inoculated into the flanks of 4-to 5-week-old athymic nude mice (Shanghai Laboratory Animal Company, Shanghai, China) subcutaneously to generate a subcutaneous xenograft tumor model. After 2 weeks, the tumor model was successfully constructed, the mice were treated single and combined with 100 mg/kg RSL3 (2 times/week) and 1.0 g/kg/days PF. Tumor volumes were measured every 4 days to draw the growth curve. Mice were sacrificed 4 weeks after cell injection. Tumor xenografts were collected, photographed, and weighed and the tumor apoptosis was analyzed by Tunel staining.
Click to Show/Hide
|
||||
Response regulation | Paeoniflorin (PF) can function as an antitumor agent for glioma treatment by targeting NEDD4L-dependent STAT3 ubiquitination as well as by regulating the Nrf2/GPX4 signaling axis, which might trigger ferroptosis. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [3] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | ALZ003 | Investigative | |||
Responsed Regulator | Androgen receptor (AR) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Apoptosis | hsa04210 | ||||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 | |
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
In Vivo Model |
NOD-SCID male mice (8-week-old) were purchased from BioLASCO Taiwan Co., Ltd. (Taipei, Taiwan). For glioblastoma and TMZ-resistant glioblastoma transplantation, luciferase-expressed U87MG cells (2 x 105) and U87MG-R cells (2 x 105) were injected into the cortex, respectively, at the depth of 3 mm using stereotactic guidance and microprocessor single syringe (Harvard Apparatus, Holliston, MA, USA). After 10 days of transplantation, TMZ (15 mg/kg) and ALZ003 were orally and intravenously administrated three times per week, respectively.
Click to Show/Hide
|
||||
Response regulation | ALZ003 targeting AR for degradation strongly exhibits the therapeutic effect on glioblastoma, including TMZ-resistant tumor,in vitroandin vivo. Particularly, GPX4 was positively regulated by AR, and overexpression of AR also prevented lipid peroxidation. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 ( MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 (MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [4] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Responsed Regulator | Endoplasmic reticulum chaperone BiP (HSPA5) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U-373MG cells | Astrocytoma | Homo sapiens | CVCL_2219 | ||
HT22 cells | Normal | Mus musculus | CVCL_0321 | ||
In Vivo Model |
Specific pathogen-free athymic nude BALB/c mice (4-6 weeks old) were obtained from Guangdong Experimental Animal Centre (Guangzhou, China). To generate murine subcutaneous tumors, cells (for U251: 2 x 106 cells; for U373: 2 x 106 cells) were suspended in 0.2 ml PBS and injected into the flanks of mice (n = 6/group). Tumor volume was measured once every 3 days using calipers.
Click to Show/Hide
|
||||
Response regulation | HSPA5 upregulation increased the expression and activity of glutathione peroxidase 4 (GPX4), which neutralized Dihydroartemisinin-induced lipid peroxidation and thus protected glioma cells from ferroptosis. Ferroptosis might be a novel anticancer mechanism of DHA in glioma and HSPA5 may serve as a negative regulator of DHA-induced ferroptosis. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [4] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Responsed Regulator | Endoplasmic reticulum chaperone BiP (HSPA5) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U-373MG cells | Astrocytoma | Homo sapiens | CVCL_2219 | ||
HT22 cells | Normal | Mus musculus | CVCL_0321 | ||
In Vivo Model |
Specific pathogen-free athymic nude BALB/c mice (4-6 weeks old) were obtained from Guangdong Experimental Animal Centre (Guangzhou, China). To generate murine subcutaneous tumors, cells (for U251: 2 x 106 cells; for U373: 2 x 106 cells) were suspended in 0.2 ml PBS and injected into the flanks of mice (n = 6/group). Tumor volume was measured once every 3 days using calipers.
Click to Show/Hide
|
||||
Response regulation | HSPA5 upregulation increased the expression and activity of glutathione peroxidase 4 (GPX4), which neutralized Dihydroartemisinin-induced lipid peroxidation and thus protected glioma cells from ferroptosis. Ferroptosis might be a novel anticancer mechanism of DHA in glioma and HSPA5 may serve as a negative regulator of DHA-induced ferroptosis. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Fatostatin | Investigative | |||
Responsed Regulator | RAC-alpha serine/threonine-protein kinase (AKT1) | Suppressor | |||
Pathway Response | Cell adhesion molecules | hsa04514 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
After anesthetizing the nude mice with isoflurane inhalation, we injected 1 x 106 U87 cells that were engineered for the expression of luciferase into the right striatum (3.5 mm from the midline of the brain and 2 mm in front of the coronal suture, injection depth of 3 mm from the brain surface) of the nude mice to establish an intracranial xenograft model. For the detection of pharmacokinetics in mice, RhoB-loaded p28-PLGA NPs were injected into the mice (n = 3) through the tail vein. We collected blood samples at predetermined time points, quantified the RhoB concentrations, and plotted them with time. To characterize NPs for GBM treatment, we randomly divided the tumor-bearing mice into four groups (n = 8) treated with PBS, free fatostatin (25 mg/kg), NPs-FAT (fatostatin equivalent dose at 25 mg/kg), and p28-NPs-FAT (fatostatin equivalent dose at 25 mg/kg). After 7 days of tumor inoculation, the treatment was conducted 3 days per week for 4 weeks. In addition, we performed IVIS imaging of intracranial tumors at 1, 3, and 5 weeks after tumor inoculation to observe tumor progression. IVIS was also used to carry out imaging of IR780-loaded NPs. The mice were monitored regularly and euthanized when they exhibited severe neurological symptoms and/or obvious weight loss (>20% of their body weight). We sacrificed a separate cohort of mice five weeks after tumor inoculation for pathological staining (n = 3).
Click to Show/Hide
|
||||
Response regulation | Fatostatin induces ferroptosis by inhibiting the AKT/mTORC1/GPX4 signaling pathway in glioblastoma. In addition, fatostatin inhibits cell proliferation and the EMT process through the AKT/mTORC1 signaling pathway. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Fatostatin | Investigative | |||
Responsed Regulator | Serine/threonine-protein kinase mTOR (MTOR) | Suppressor | |||
Pathway Response | Cell adhesion molecules | hsa04514 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
After anesthetizing the nude mice with isoflurane inhalation, we injected 1 x 106 U87 cells that were engineered for the expression of luciferase into the right striatum (3.5 mm from the midline of the brain and 2 mm in front of the coronal suture, injection depth of 3 mm from the brain surface) of the nude mice to establish an intracranial xenograft model. For the detection of pharmacokinetics in mice, RhoB-loaded p28-PLGA NPs were injected into the mice (n = 3) through the tail vein. We collected blood samples at predetermined time points, quantified the RhoB concentrations, and plotted them with time. To characterize NPs for GBM treatment, we randomly divided the tumor-bearing mice into four groups (n = 8) treated with PBS, free fatostatin (25 mg/kg), NPs-FAT (fatostatin equivalent dose at 25 mg/kg), and p28-NPs-FAT (fatostatin equivalent dose at 25 mg/kg). After 7 days of tumor inoculation, the treatment was conducted 3 days per week for 4 weeks. In addition, we performed IVIS imaging of intracranial tumors at 1, 3, and 5 weeks after tumor inoculation to observe tumor progression. IVIS was also used to carry out imaging of IR780-loaded NPs. The mice were monitored regularly and euthanized when they exhibited severe neurological symptoms and/or obvious weight loss (>20% of their body weight). We sacrificed a separate cohort of mice five weeks after tumor inoculation for pathological staining (n = 3).
Click to Show/Hide
|
||||
Response regulation | Fatostatin induces ferroptosis by inhibiting the AKT/ mTORC1/GPX4 signaling pathway in glioblastoma. In addition, fatostatin inhibits cell proliferation and the EMT process through the AKT/mTORC1 signaling pathway. | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [5] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Fatostatin | Investigative | |||
Responsed Regulator | Serine/threonine-protein kinase mTOR (MTOR) | Suppressor | |||
Pathway Response | Cell adhesion molecules | hsa04514 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
After anesthetizing the nude mice with isoflurane inhalation, we injected 1 x 106 U87 cells that were engineered for the expression of luciferase into the right striatum (3.5 mm from the midline of the brain and 2 mm in front of the coronal suture, injection depth of 3 mm from the brain surface) of the nude mice to establish an intracranial xenograft model. For the detection of pharmacokinetics in mice, RhoB-loaded p28-PLGA NPs were injected into the mice (n = 3) through the tail vein. We collected blood samples at predetermined time points, quantified the RhoB concentrations, and plotted them with time. To characterize NPs for GBM treatment, we randomly divided the tumor-bearing mice into four groups (n = 8) treated with PBS, free fatostatin (25 mg/kg), NPs-FAT (fatostatin equivalent dose at 25 mg/kg), and p28-NPs-FAT (fatostatin equivalent dose at 25 mg/kg). After 7 days of tumor inoculation, the treatment was conducted 3 days per week for 4 weeks. In addition, we performed IVIS imaging of intracranial tumors at 1, 3, and 5 weeks after tumor inoculation to observe tumor progression. IVIS was also used to carry out imaging of IR780-loaded NPs. The mice were monitored regularly and euthanized when they exhibited severe neurological symptoms and/or obvious weight loss (>20% of their body weight). We sacrificed a separate cohort of mice five weeks after tumor inoculation for pathological staining (n = 3).
Click to Show/Hide
|
||||
Response regulation | Fatostatin induces ferroptosis by inhibiting the AKT/mTORC1/GPX4 signaling pathway in glioblastoma. In addition, fatostatin inhibits cell proliferation and the EMT process through the AKT/mTORC1 signaling pathway. | ||||
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target | [10] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | RSL3 | Investigative | |||
Pathway Response | NF-kappa B signaling pathway | hsa04064 | |||
Fatty acid metabolism | hsa01212 | ||||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Female B-NDG mice (4-6 weeks old, 16-20 g) were purchased from Biocytogen (Biocytogen Jiangsu Co., Ltd., Jiangsu, China) and housed under specific pathogen-free conditions. 5 x 106 U87 cells were resuspended in 200 uL PBS buffer and then inoculated into the left hind limb of each mouse. Once tumor volumes reached >=50 mm3, the mice were randomly divided into four groups (n = 5): the control, RSL3-only, BAY-only, and RSL3 plus BAY groups. Chemicals were administered through intratumor injection (100 mg/kg for RSL3 and 1 mg/kg for BAY 11-7082) biweekly for two weeks.
Click to Show/Hide
|
||||
Response regulation | NF-kB pathway activation is vital for RSL3-induced ferroptosis in glioblastoma cells both in vitro and in vivo. Furthermore, RNAi-mediated GPX4 silencing cannot trigger ferroptosis in glioblastoma cells unless the NF-kB pathway is activated simultaneously. Finally, NF-kB pathway activation promotes ferroptosis by downregulating the expression of ATF4 and SLC7A11. | ||||
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target | [15] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
Response regulation | Dihydroartemisinin (DHA) had a selective killing effect on glioblastoma, which was associated with over-expression of transferrin receptors. The primary mechanism by which DHA caused ferroptosis was down-regulation of GPX4 and the following lipid ROS accumulation. | ||||
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target | [9] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
All BALB/C nude mice were purchased from Huafukang Biotechnology (Beijing, China). These mice were 5 weeks old and weighed 14-16 g. We established subcutaneous tumour-forming mouse model by injecting 5 x 106 U87 cells into the lateral abdomen of BALB/C nude mice. Animals were then treated with DHA solvent (50 mg/kg) by intragastric administration once a day for 26 days.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin could promote ferroptosis in glioma cells. Low expression of GPX4 and high expression of HMOX1 were identified in DHA treated glioma cells. MAZ was further identified as the direct target of long noncoding RNA (lncRNA) TUG1 through luciferase assay. Downregulated expression of TUG1 and upregulated expression of MAZ were identified in DHA treated glioma cells. TUG1 overexpression or inhibition of FTH1 expression could enhance the antiglioma effect of DHA in vitro and in vivo, providing a promising strategy to enhance the antitumor effect of DHA in glioma. | ||||
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target | [16] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydrotanshinone I | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | HEB (Human glial cells) | ||||
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
Response regulation | Dihydrotanshinone I (DHI) inhibited the proliferation of human glioma cells. Following treatment of the U251 and U87 cells with DHI, changes in the expression levels of ferroptosis-associated proteins were observed; the expression level of GPX4 decreased and that of ACSL-4 increased. DHI also increased the levels of LDH and MDA in the human glioma cells and reduced the GSH/GSSG ratio. | ||||
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target | [17] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Sodium Selenite | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 | |
HeLa cells | Endocervical adenocarcinoma | Homo sapiens | CVCL_0030 | ||
MCF-7 cells | Breast carcinoma | Homo sapiens | CVCL_0031 | ||
PC-3 cells | Prostate carcinoma | Homo sapiens | CVCL_0035 | ||
HT-29 cells | Colon adenocarcinoma | Homo sapiens | CVCL_0320 | ||
SVG p12 cells | Normal | Homo sapiens | CVCL_3797 | ||
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
HT-1080 cells | Fibrosarcoma | Homo sapiens | CVCL_0317 | ||
HCT 116 cells | Colon carcinoma | Homo sapiens | CVCL_0291 | ||
Response regulation | Sodium selenite (SS) down-regulates ferroptosis regulators; solute carrier family 7 member 11 (SLC7A11), glutathione (GSH), and glutathione peroxidase 4 (GPx4), while it up-regulates iron accumulation and lipid peroxidation (LPO) in Glioblastoma. | ||||
Nuclear receptor coactivator 4 (NCOA4)
In total 3 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 ( MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 (MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [18] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Regulator | Coatomer subunit zeta-1 (COPZ1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
Cell proliferation | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | ||
T98 cells | Glioblastoma | Homo sapiens | CVCL_B368 | ||
In Vivo Model |
Mice were divided into two groups (10 mice per group) and anesthetized with an intraperitoneal injection (80 uL) containing ketamine HCl (25 mg/mL), xylazine (2.5 mg/mL), and 14.25% ethyl alcohol (diluted 1:3 in 0.9% NaCl). U87MG-NC and U87MG-sh-COPZ1#1 glioma cells (106 cells diluted in 10 uL PBS per animal) were injected into the right frontal lobes of each mouse using the following coordinates: 1 mm anterior and 2.5 mm lateral to the bregma, at a depth of 2 mm.
Click to Show/Hide
|
||||
Response regulation | COPZ1 knockdown also led to the increase in nuclear receptor coactivator 4 (NCOA4), resulting in the degradation of ferritin, and a subsequent increase in the intracellular levels of ferrous iron and ultimately ferroptosis.The COPZ1/NCOA4/FTH1 axis is therefore a novel therapeutic target for the treatment of human glioblastoma. | ||||
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 | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Peoniflorin | Investigative | |||
Responsed Regulator | E3 ubiquitin-protein ligase NEDD4-like (NEDD4L) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
In Vivo Model |
U251 cells (6 x 106) were inoculated into the flanks of 4-to 5-week-old athymic nude mice (Shanghai Laboratory Animal Company, Shanghai, China) subcutaneously to generate a subcutaneous xenograft tumor model. After 2 weeks, the tumor model was successfully constructed, the mice were treated single and combined with 100 mg/kg RSL3 (2 times/week) and 1.0 g/kg/days PF. Tumor volumes were measured every 4 days to draw the growth curve. Mice were sacrificed 4 weeks after cell injection. Tumor xenografts were collected, photographed, and weighed and the tumor apoptosis was analyzed by Tunel staining.
Click to Show/Hide
|
||||
Response regulation | Paeoniflorin (PF) can function as an antitumor agent for glioma treatment by targeting NEDD4L-dependent STAT3 ubiquitination as well as by regulating the Nrf2/GPX4 signaling axis, which might trigger ferroptosis. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [2] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Peoniflorin | Investigative | |||
Responsed Regulator | Signal transducer and activator of transcription 3 (STAT3) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Ubiquitin mediated proteolysis | hsa04120 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
In Vivo Model |
U251 cells (6 x 106) were inoculated into the flanks of 4-to 5-week-old athymic nude mice (Shanghai Laboratory Animal Company, Shanghai, China) subcutaneously to generate a subcutaneous xenograft tumor model. After 2 weeks, the tumor model was successfully constructed, the mice were treated single and combined with 100 mg/kg RSL3 (2 times/week) and 1.0 g/kg/days PF. Tumor volumes were measured every 4 days to draw the growth curve. Mice were sacrificed 4 weeks after cell injection. Tumor xenografts were collected, photographed, and weighed and the tumor apoptosis was analyzed by Tunel staining.
Click to Show/Hide
|
||||
Response regulation | Paeoniflorin (PF) can function as an antitumor agent for glioma treatment by targeting NEDD4L-dependent STAT3 ubiquitination as well as by regulating the Nrf2/GPX4 signaling axis, which might trigger ferroptosis. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [6] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Apatinib | Investigative | |||
Responsed Regulator | Vascular endothelial growth factor receptor 2 (KDR) | Driver | |||
Pathway Response | Pathways in cancer | hsa05200 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Female BALB/c nude mice (age, 4 weeks old) were purchased from Changzhou Cavens Experimental Animal Co., Ltd. (Changzhou, China).The gliomas from the nude mice were fixed in 10% paraformaldehyde at 4 for 12 h and then dehydrated in different concentrations of ethanol. The tumor tissues were permeabilized using xylene and embedded in paraffin. They were then sliced (0.5 um), rehydrated, and stained with HE at 4 for 10 min and sealed. For IHC assessment of Ki-67 in gliomas, the DAKO Envision system (Dako; Agilent Technologies, Inc.) was used.
Click to Show/Hide
|
||||
Response regulation | Apatinib could restrain proliferation of glioma cells through induction of ferroptosis via inhibiting the activation of VEGFR2/Nrf2/Keap1 pathway. Overexpression of Nrf2 could counteract the induction of ferroptosis by apatinib. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [6] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Apatinib | Investigative | |||
Responsed Regulator | Kelch-like ECH-associated protein 1 (KEAP1) | Driver | |||
Pathway Response | Pathways in cancer | hsa05200 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Female BALB/c nude mice (age, 4 weeks old) were purchased from Changzhou Cavens Experimental Animal Co., Ltd. (Changzhou, China).The gliomas from the nude mice were fixed in 10% paraformaldehyde at 4 for 12 h and then dehydrated in different concentrations of ethanol. The tumor tissues were permeabilized using xylene and embedded in paraffin. They were then sliced (0.5 um), rehydrated, and stained with HE at 4 for 10 min and sealed. For IHC assessment of Ki-67 in gliomas, the DAKO Envision system (Dako; Agilent Technologies, Inc.) was used.
Click to Show/Hide
|
||||
Response regulation | Apatinib could restrain proliferation of glioma cells through induction of ferroptosis via inhibiting the activation of VEGFR2/Nrf2/ Keap1 pathway. Overexpression of Nrf2 could counteract the induction of ferroptosis by apatinib. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [6] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Apatinib | Investigative | |||
Responsed Regulator | Kelch-like ECH-associated protein 1 (KEAP1) | Driver | |||
Pathway Response | Pathways in cancer | hsa05200 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Female BALB/c nude mice (age, 4 weeks old) were purchased from Changzhou Cavens Experimental Animal Co., Ltd. (Changzhou, China).The gliomas from the nude mice were fixed in 10% paraformaldehyde at 4 for 12 h and then dehydrated in different concentrations of ethanol. The tumor tissues were permeabilized using xylene and embedded in paraffin. They were then sliced (0.5 um), rehydrated, and stained with HE at 4 for 10 min and sealed. For IHC assessment of Ki-67 in gliomas, the DAKO Envision system (Dako; Agilent Technologies, Inc.) was used.
Click to Show/Hide
|
||||
Response regulation | Apatinib could restrain proliferation of glioma cells through induction of ferroptosis via inhibiting the activation of VEGFR2/Nrf2/Keap1 pathway. Overexpression of Nrf2 could counteract the induction of ferroptosis by apatinib. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [19] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Ibuprofen | Approved | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
In the intracranial glioma model, U87MG cells (5 x 105) were intracerebrally injected into the left side (bregma: 1 mm; lateral: 2 mm; ventral: 3 mm) of the brains of nude mice. Two weeks after tumor cell transplantation, mouse brains were scanned to detect tumor formation using a 3.0-T scanner (GE Signa HD MRI Systems). Then, mice were divided randomly into two groups (n = 6/group) and treated with vehicle control (PBS), oribuprofen (20 mg/kg), in 100 ul of PBS given i.p. 1x/day, 5 days/week.
Click to Show/Hide
|
||||
Response regulation | Ibuprofen could induce ferroptosis of glioblastoma cells via downregulation of Nrf2 signaling pathway and is a potential drug for glioma treatment. All the data suggested that Nrf2 could regulate the expression of GPX4 and SLC7A11 in glioma cells. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [20] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Mitogen-activated protein kinase 8 (MAPK8) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
hACs (Normal human astrocyte cells) | |||||
In Vivo Model |
BALB/c nude mice (female, four-week-old) were purchased from the Nanjing Medical University Experimental Animal Department. Female mice were randomly divided into test group and control group. 2.5 x 105 LN229/TMZ cells transfected with sh-MAPK8-1 or sh-LINC01564-1 were injected into the brain of mice in test group, taking the mice injected with sh-NC-transfected ones as control. Seven days later, the mice were treated with TMZ (66 mg/kg per day, 5 days/cycle, 4 cycles in total) as a monotherapy. Tumor volume was monitored every three days in the period of TMZ treatment. The mice were killed 28 days after the injection. Tumors were excised from mice for observation and weighing as well as the detection of the level of ROS, iron (Fe2+) and proteins (i.e., NFE2L2, NQO1, FTH1 and HO-1).
Click to Show/Hide
|
||||
Response regulation | LINC01564 promotes the temozolomide (TMZ) resistance of glioma cells by upregulating NFE2L2 expression to inhibit ferroptosis. LINC01564 promotes MAPK8 mRNA stability by recruiting SRSF1, and MAPK8 was positively correlated with NFE2L2 and its targets, proving its mediation of NFE2L2. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [20] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Serine/arginine-rich splicing factor 1 (SRSF1) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
hACs (Normal human astrocyte cells) | |||||
In Vivo Model |
BALB/c nude mice (female, four-week-old) were purchased from the Nanjing Medical University Experimental Animal Department. Female mice were randomly divided into test group and control group. 2.5 x 105 LN229/TMZ cells transfected with sh-MAPK8-1 or sh-LINC01564-1 were injected into the brain of mice in test group, taking the mice injected with sh-NC-transfected ones as control. Seven days later, the mice were treated with TMZ (66 mg/kg per day, 5 days/cycle, 4 cycles in total) as a monotherapy. Tumor volume was monitored every three days in the period of TMZ treatment. The mice were killed 28 days after the injection. Tumors were excised from mice for observation and weighing as well as the detection of the level of ROS, iron (Fe2+) and proteins (i.e., NFE2L2, NQO1, FTH1 and HO-1).
Click to Show/Hide
|
||||
Response regulation | LINC01564 promotes the temozolomide (TMZ) resistance of glioma cells by upregulating NFE2L2 expression to inhibit ferroptosis. LINC01564 promotes MAPK8 mRNA stability by recruiting SRSF1, and MAPK8 was positively correlated with NFE2L2 and its targets, proving its mediation of NFE2L2. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [20] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | LINC01564 (IncRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
hACs (Normal human astrocyte cells) | |||||
In Vivo Model |
BALB/c nude mice (female, four-week-old) were purchased from the Nanjing Medical University Experimental Animal Department. Female mice were randomly divided into test group and control group. 2.5 x 105 LN229/TMZ cells transfected with sh-MAPK8-1 or sh-LINC01564-1 were injected into the brain of mice in test group, taking the mice injected with sh-NC-transfected ones as control. Seven days later, the mice were treated with TMZ (66 mg/kg per day, 5 days/cycle, 4 cycles in total) as a monotherapy. Tumor volume was monitored every three days in the period of TMZ treatment. The mice were killed 28 days after the injection. Tumors were excised from mice for observation and weighing as well as the detection of the level of ROS, iron (Fe2+) and proteins (i.e., NFE2L2, NQO1, FTH1 and HO-1).
Click to Show/Hide
|
||||
Response regulation | LINC01564 promotes the temozolomide (TMZ) resistance of glioma cells by upregulating NFE2L2 expression to inhibit ferroptosis. LINC01564 promotes MAPK8 mRNA stability by recruiting SRSF1, and MAPK8 was positively correlated with NFE2L2 and its targets, proving its mediation of NFE2L2. | ||||
Natural resistance-associated macrophage protein 2 (SLC11A2)
In total 3 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 ( MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [1] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Artesunate | Investigative | |||
Responsed Regulator | Mitogen-activated protein kinase 14 (MAPK14) | Driver | |||
Pathway Response | MAPK signaling pathway | hsa04010 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
In Vivo Model |
The xenografts were established via the subcutaneous inoculation of U251 cells (1 x 107 cells/per mouse) into the armpit of one mouse. After two weeks of growth, the cancer tissues were cut into pieces with the dimensions of 1.5 x 1.5 x 1.5 mm3 and inoculated subcutaneously into the right armpit of the mice with a puncture needle. When tumor volume reached approximately 80 mm3, mice were randomly divided into four groups (n = 5): Vehicle control, ART (20 mg/kg), ART (40 mg/kg), and TMZ (40 mg/kg). TMZ was used as the positive control. Drugs and vehicle were given by intraperitoneal injection daily for 21 days. Tumor volume and body weight were measured every three days.
Click to Show/Hide
|
||||
Response regulation | Artesunate triggers ferroptosis in glioblastoma in vitro and in vivo through regulation of iron metabolism and p38 (MAPK14) and ERK signaling pathways. Meanwhile, ART reduced the protein level of GPX4 and FPN1, increased the protein level of DMT1, TfR, ferritin and NCOA4. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [21] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Temozolomide | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | TG905 (Human glioblastoma cells) | ||||
Response regulation | Temozolomide may suppress cell growth partly by inducing ferroptosis by targeting DMT1 expression in glioblastoma cells. The results also showed that temozolomide-induced ferroptosis is associated with regulation of the Nrf2/HO-1 pathway. | ||||
NADPH oxidase 4 (NOX4)
In total 2 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [7] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Brucine | Investigative | |||
Responsed Regulator | Cyclic AMP-dependent transcription factor ATF-3 (ATF3) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U118 cells | Astrocytoma | Homo sapiens | CVCL_0633 | |
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
In Vivo Model |
The athymic BALB/c nude mice (4 weeks; 20-22 g; Beijing Vital River Laboratory Animal Technology Company, China) were housed in a specific pathogen-free environment under a 12-h lightdark cycle with free access to food and water. The animals were allowed to acclimatize to their surroundings for 3 days. U87 cells (1 x 106) in the logarithmic growth phase in 100 uL PBS were subcutaneously injected into the right flank. Therapeutic experiments were started when the tumor reached around 150 mm3 after about 10 days. Mice were allocated to receive intraperitoneal injections of vehicle (control group, n = 6) or 40 mg/kg bodyweight (n = 6) in the same volume (50 uL) once a day for 13 times.
Click to Show/Hide
|
||||
Response regulation | Brucine inhibited glioma cell growth in vitro and in vivo, and brucine induced ATF3 upregulation and translocation into nuclei via activation of ER stress. ATF3 promoted brucine-induced H2O2 accumulation via upregulating NOX4 and SOD1 to generate H2O2 on one hand, and downregulating catalase and xCT to prevent H2O2 degradation on the other hand. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [11] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Pseudolaric acid B | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_0022 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
SHG-44 cells | Astrocytoma | Homo sapiens | CVCL_6728 | ||
In Vivo Model |
Twenty athymic BALB/c nude mice (aged 4 weeks, weight 20-22 g, from Shanghai laboratory animal Center, Shanghai, China) were housed in a specific pathogen-free environment. A total of 1 x 106 logarithmically growing C6 cells in 100 uL of PBS were subcutaneously injected into the right flank of each mouse. Therapeutic experiments were started when the tumor reached about 150 mm3 after about 7 days. The mice were allocated to receive intraperitoneal injections of vehicle (control group, n = 5/group), PAB at the dosage of 10 mg/kg body weight (n = 10/group) and 20 mg/kg body weight (n = 10/group) in the same volume 50 uL once a days for 8 times.
Click to Show/Hide
|
||||
Response regulation | Pseudolaric acid B (PAB) improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT pathway, which further exacerbated accumulation of H2O2and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment. | ||||
Ferritin heavy chain (FTH1)
In total 5 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Amentoflavone | Investigative | |||
Responsed Regulator | Microtubule-associated proteins 1A/1B light chain 3B {ECO:0000305} (MAP1LC3B) | Driver | |||
Pathway Response | mTOR signaling pathway | hsa04150 | |||
Fatty acid metabolism | hsa01212 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U-373MG cells | Astrocytoma | Homo sapiens | CVCL_2219 | ||
In Vivo Model |
1 x 107 U251 cells were subcutaneously injected into the right back of the four-week-old BALB/c nude mice. After the tumor grown to 100 mm3, mice were randomly divided into 3 groups: mice in control group receivedintraperitoneal injectionof saline, while mice in AF treatment group received intraperitoneal injection at dosages of 40 mg/kg/day or 80 mg/kg/day, respectively.
Click to Show/Hide
|
||||
Response regulation | Amentoflavone (AF) triggered ferroptosis in autophagy-dependent manner. Furthermore, the expression of LC3B, Beclin1, ATG5, ATG7 were increased, and the expression of FTH were decreased by AF in a dose-dependent manner in vivo. AF has the potential to be considered as a novel treatment agent in glioma. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Amentoflavone | Investigative | |||
Responsed Regulator | Autophagy protein 5 (ATG5) | Driver | |||
Pathway Response | mTOR signaling pathway | hsa04150 | |||
Fatty acid metabolism | hsa01212 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U-373MG cells | Astrocytoma | Homo sapiens | CVCL_2219 | ||
In Vivo Model |
1 x 107 U251 cells were subcutaneously injected into the right back of the four-week-old BALB/c nude mice. After the tumor grown to 100 mm3, mice were randomly divided into 3 groups: mice in control group receivedintraperitoneal injectionof saline, while mice in AF treatment group received intraperitoneal injection at dosages of 40 mg/kg/day or 80 mg/kg/day, respectively.
Click to Show/Hide
|
||||
Response regulation | Amentoflavone (AF) triggered ferroptosis in autophagy-dependent manner. Furthermore, the expression of LC3B, Beclin1, ATG5, ATG7 were increased, and the expression of FTH were decreased by AF in a dose-dependent manner in vivo. AF has the potential to be considered as a novel treatment agent in glioma. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Marker/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Amentoflavone | Investigative | |||
Responsed Regulator | Ubiquitin-like modifier-activating enzyme ATG7 (ATG7) | Driver | |||
Pathway Response | mTOR signaling pathway | hsa04150 | |||
Fatty acid metabolism | hsa01212 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U-373MG cells | Astrocytoma | Homo sapiens | CVCL_2219 | ||
In Vivo Model |
1 x 107 U251 cells were subcutaneously injected into the right back of the four-week-old BALB/c nude mice. After the tumor grown to 100 mm3, mice were randomly divided into 3 groups: mice in control group receivedintraperitoneal injectionof saline, while mice in AF treatment group received intraperitoneal injection at dosages of 40 mg/kg/day or 80 mg/kg/day, respectively.
Click to Show/Hide
|
||||
Response regulation | Amentoflavone (AF) triggered ferroptosis in autophagy-dependent manner. Furthermore, the expression of LC3B, Beclin1, ATG5, ATG7 were increased, and the expression of FTH were decreased by AF in a dose-dependent manner in vivo. AF has the potential to be considered as a novel treatment agent in glioma. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [9] | ||||
Target for Ferroptosis | Marker | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Responsed Regulator | Myc-associated zinc finger protein (MAZ) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
All BALB/C nude mice were purchased from Huafukang Biotechnology (Beijing, China). These mice were 5 weeks old and weighed 14-16 g. We established subcutaneous tumour-forming mouse model by injecting 5 x 106 U87 cells into the lateral abdomen of BALB/C nude mice. Animals were then treated with DHA solvent (50 mg/kg) by intragastric administration once a day for 26 days.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin (DHA) could promote ferroptosis in glioma cells. Low expression of GPX4 and high expression of HMOX1 were identified in DHA treated glioma cells. MAZ was further identified as the direct target of long noncoding RNA (lncRNA) TUG1 through luciferase assay. Downregulated expression of TUG1 and upregulated expression of MAZ were identified in DHA treated glioma cells. TUG1 overexpression or inhibition of FTH1 expression could enhance the antiglioma effect of DHA in vitro and in vivo, providing a promising strategy to enhance the antitumor effect of DHA in glioma. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [9] | ||||
Target for Ferroptosis | Marker | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Responsed Regulator | TUG1 (IncRNA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
All BALB/C nude mice were purchased from Huafukang Biotechnology (Beijing, China). These mice were 5 weeks old and weighed 14-16 g. We established subcutaneous tumour-forming mouse model by injecting 5 x 106 U87 cells into the lateral abdomen of BALB/C nude mice. Animals were then treated with DHA solvent (50 mg/kg) by intragastric administration once a day for 26 days.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin (DHA) could promote ferroptosis in glioma cells. Low expression of GPX4 and high expression of HMOX1 were identified in DHA treated glioma cells. MAZ was further identified as the direct target of long noncoding RNA (lncRNA) TUG1 through luciferase assay. Downregulated expression of TUG1 and upregulated expression of MAZ were identified in DHA treated glioma cells. TUG1 overexpression or inhibition of FTH1 expression could enhance the antiglioma effect of DHA in vitro and in vivo, providing a promising strategy to enhance the antitumor effect of DHA in glioma. | ||||
Cystine/glutamate transporter (SLC7A11)
In total 6 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [7] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Brucine | Investigative | |||
Responsed Regulator | Cyclic AMP-dependent transcription factor ATF-3 (ATF3) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U118 cells | Astrocytoma | Homo sapiens | CVCL_0633 | |
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
In Vivo Model |
The athymic BALB/c nude mice (4 weeks; 20-22 g; Beijing Vital River Laboratory Animal Technology Company, China) were housed in a specific pathogen-free environment under a 12-h lightdark cycle with free access to food and water. The animals were allowed to acclimatize to their surroundings for 3 days. U87 cells (1 x 106) in the logarithmic growth phase in 100 uL PBS were subcutaneously injected into the right flank. Therapeutic experiments were started when the tumor reached around 150 mm3 after about 10 days. Mice were allocated to receive intraperitoneal injections of vehicle (control group, n = 6) or 40 mg/kg bodyweight (n = 6) in the same volume (50 uL) once a day for 13 times.
Click to Show/Hide
|
||||
Response regulation | Brucine inhibited glioma cell growth in vitro and in vivo, and brucine induced ATF3 upregulation and translocation into nuclei via activation of ER stress. ATF3 promoted brucine-induced H2O2 accumulation via upregulating NOX4 and SOD1 to generate H2O2 on one hand, and downregulating catalase and xCT (SLC7A11) to prevent H2O2 degradation on the other hand. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [10] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | RSL3 | Investigative | |||
Responsed Regulator | NF-kappa-B inhibitor alpha (NFKBIA) | Driver | |||
Pathway Response | NF-kappa B signaling pathway | hsa04064 | |||
Fatty acid metabolism | hsa01212 | ||||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Female B-NDG mice (4-6 weeks old, 16-20 g) were purchased from Biocytogen (Biocytogen Jiangsu Co., Ltd., Jiangsu, China) and housed under specific pathogen-free conditions. 5 x 106 U87 cells were resuspended in 200 uL PBS buffer and then inoculated into the left hind limb of each mouse. Once tumor volumes reached >=50 mm3, the mice were randomly divided into four groups (n = 5): the control, RSL3-only, BAY-only, and RSL3 plus BAY groups. Chemicals were administered through intratumor injection (100 mg/kg for RSL3 and 1 mg/kg for BAY 11-7082) biweekly for two weeks.
Click to Show/Hide
|
||||
Response regulation | NF-kB pathway activation is vital for RSL3-induced ferroptosis in glioblastoma cells both in vitro and in vivo. Furthermore, RNAi-mediated GPX4 silencing cannot trigger ferroptosis in glioblastoma cells unless the NF-kB pathway is activated simultaneously. Finally, NF-kB pathway activation promotes ferroptosis by downregulating the expression of ATF4 and SLC7A11. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [11] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Pseudolaric acid B | Investigative | |||
Responsed Regulator | Cellular tumor antigen p53 (TP53) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_0022 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
SHG-44 cells | Astrocytoma | Homo sapiens | CVCL_6728 | ||
In Vivo Model |
Twenty athymic BALB/c nude mice (aged 4 weeks, weight 20-22 g, from Shanghai laboratory animal Center, Shanghai, China) were housed in a specific pathogen-free environment. A total of 1 x 106 logarithmically growing C6 cells in 100 uL of PBS were subcutaneously injected into the right flank of each mouse. Therapeutic experiments were started when the tumor reached about 150 mm3 after about 7 days. The mice were allocated to receive intraperitoneal injections of vehicle (control group, n = 5/group), PAB at the dosage of 10 mg/kg body weight (n = 10/group) and 20 mg/kg body weight (n = 10/group) in the same volume 50 uL once a days for 8 times.
Click to Show/Hide
|
||||
Response regulation | Pseudolaric acid B (PAB) improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT (SLC7A11) pathway, which further exacerbated accumulation of H2O2and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Platelet-derived growth factor receptor alpha (PDGFRA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | HEB (Human glial cells) | ||||
SF126 cells | Glioblastoma | Homo sapiens | CVCL_1688 | ||
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
4-week-old female BALB/c nude mice were acquired from the National Laboratory Animal Center (Shanghai, China). Overall, 10 mice (n = 5 each group) were implanted with U251 cells stably knockdown circCDK14 by lentiviruses carrying sh-circCDK14 (Wanleibio, China), or control U251 cells with lentiviruses carrying sh-NC (Wanleibio, China). 5 x 106 cells were resuspended in phosphatebuffered saline (100 ul) and Matrigel substrate (100 ul) and injected into the right flank of nude mice. Tumor volume was documented once 7 days after implantation.
Click to Show/Hide
|
||||
Response regulation | CircCDK14 promotes the migration, invasion and proliferation of glioma cells in vitroas well as tumorigenesisin vivo. An evaluation of the underlying mechanism revealed that circCDK14 sponged miR-3938 to upregulate oncogenic gene PDGFRA expression. After knockdown of circCDK14 in glioma cells, protein levels of SLC7A11 and GPX4 decreased significantly and Fp became more sensitivity. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | hsa-miR-3938 (miRNA) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | HEB (Human glial cells) | ||||
SF126 cells | Glioblastoma | Homo sapiens | CVCL_1688 | ||
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
4-week-old female BALB/c nude mice were acquired from the National Laboratory Animal Center (Shanghai, China). Overall, 10 mice (n = 5 each group) were implanted with U251 cells stably knockdown circCDK14 by lentiviruses carrying sh-circCDK14 (Wanleibio, China), or control U251 cells with lentiviruses carrying sh-NC (Wanleibio, China). 5 x 106 cells were resuspended in phosphatebuffered saline (100 ul) and Matrigel substrate (100 ul) and injected into the right flank of nude mice. Tumor volume was documented once 7 days after implantation.
Click to Show/Hide
|
||||
Response regulation | CircCDK14 promotes the migration, invasion and proliferation of glioma cells in vitroas well as tumorigenesisin vivo. An evaluation of the underlying mechanism revealed that circCDK14 sponged miR-3938 to upregulate oncogenic gene PDGFRA expression. After knockdown of circCDK14 in glioma cells, protein levels of SLC7A11 and GPX4 decreased significantly and Fp became more sensitivity. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [27] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | CircCDK14 (circRNA) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Fatty acid metabolism | hsa01212 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | HEB (Human glial cells) | ||||
SF126 cells | Glioblastoma | Homo sapiens | CVCL_1688 | ||
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
4-week-old female BALB/c nude mice were acquired from the National Laboratory Animal Center (Shanghai, China). Overall, 10 mice (n = 5 each group) were implanted with U251 cells stably knockdown circCDK14 by lentiviruses carrying sh-circCDK14 (Wanleibio, China), or control U251 cells with lentiviruses carrying sh-NC (Wanleibio, China). 5 x 106 cells were resuspended in phosphatebuffered saline (100 ul) and Matrigel substrate (100 ul) and injected into the right flank of nude mice. Tumor volume was documented once 7 days after implantation.
Click to Show/Hide
|
||||
Response regulation | CircCDK14 promotes the migration, invasion and proliferation of glioma cells in vitroas well as tumorigenesisin vivo. An evaluation of the underlying mechanism revealed that circCDK14 sponged miR-3938 to upregulate oncogenic gene PDGFRA expression. After knockdown of circCDK14 in glioma cells, protein levels of SLC7A11 and GPX4 decreased significantly and Fp became more sensitivity. | ||||
Unspecific Target
In total 16 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [10] | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | RSL3 | Investigative | |||
Responsed Regulator | Cyclic AMP-dependent transcription factor ATF-4 (ATF4) | Suppressor | |||
Pathway Response | NF-kappa B signaling pathway | hsa04064 | |||
Fatty acid metabolism | hsa01212 | ||||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Female B-NDG mice (4-6 weeks old, 16-20 g) were purchased from Biocytogen (Biocytogen Jiangsu Co., Ltd., Jiangsu, China) and housed under specific pathogen-free conditions. 5 x 106 U87 cells were resuspended in 200 uL PBS buffer and then inoculated into the left hind limb of each mouse. Once tumor volumes reached >=50 mm3, the mice were randomly divided into four groups (n = 5): the control, RSL3-only, BAY-only, and RSL3 plus BAY groups. Chemicals were administered through intratumor injection (100 mg/kg for RSL3 and 1 mg/kg for BAY 11-7082) biweekly for two weeks.
Click to Show/Hide
|
||||
Response regulation | NF-kB pathway activation is vital for RSL3-induced ferroptosis in glioblastoma cells both in vitro and in vivo. Furthermore, RNAi-mediated GPX4 silencing cannot trigger ferroptosis in glioblastoma cells unless the NF-kB pathway is activated simultaneously. Finally, NF-kB pathway activation promotes ferroptosis by downregulating the expression of ATF4 and SLC7A11. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [12] | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Boric Acid | Investigative | |||
Responsed Regulator | Semaphorin-3F (SEMA3F) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | C6 cells | Malignant glioma | Rattus norvegicus | CVCL_0194 | |
Response regulation | Our hypothesis and results provided proof that boric acid prevent tumor progression by regulation of ferroptosis, apoptosis and semaphorin signaling pathway. BA was found to have more effective on SEMA3F/NP2 upregulated against C6 untreated cells. What's more, the novel anticancer candidate drug effect of BA were principally associated with the ACSL4/GPX4, TOS/TAS biochemical marker and SEMA3F/NP2 signaling pathways. Dose-decently BA induced oxidative and ferroptosis in Glioblastoma multiform. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [12] | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Boric Acid | Investigative | |||
Responsed Regulator | Neuronal pentraxin-2 (NPTX2) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
In Vitro Model | C6 cells | Malignant glioma | Rattus norvegicus | CVCL_0194 | |
Response regulation | Our hypothesis and results provided proof that boric acid prevent tumor progression by regulation of ferroptosis, apoptosis and semaphorin signaling pathway. BA was found to have more effective on SEMA3F/NP2 upregulated against C6 untreated cells. What's more, the novel anticancer candidate drug effect of BA were principally associated with the ACSL4/GPX4, TOS/TAS biochemical marker and SEMA3F/NP2 signaling pathways. Dose-decently BA induced oxidative and ferroptosis Glioblastoma multiform. | ||||
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target | [28] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Misonidazole | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | mGICs (Mouse glioma initiating cells) | ||||
mGSCs (Mouse glioma stem cells) | |||||
In Vivo Model |
Female C57BL/6J mice (age 6-8 weeks) were anesthetized and placed into a stereotactic apparatus (David Kopf Instruments, Tujunga, CA). One thousand viable GSC-H cells were injected into the right hemisphere at a position 2 mm lateral to the bregma and 3 mm below the brain surface. After 10 days, animals were exposed to 15 Gy (radiation dose rate, 1.45 Gy/min) with or without prior injection of doranidazole (200 mg/kg, i.p.). Radiation was confined to the brain by protection of the body with a lead shield.
Click to Show/Hide
|
||||
Response regulation | Doranidazole and misonidazole, but not metronidazole, manifested radiation-independent cytotoxicity for hypoxic glioma stem cells (GSCs) that was mediated by ferroptosis induced partially through blockade of mitochondrial complexes I and II and resultant metabolic alterations in oxidative stress responses. | ||||
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target | [28] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Doranidazole | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | mGICs (Mouse glioma initiating cells) | ||||
mGSCs (Mouse glioma stem cells) | |||||
In Vivo Model |
Female C57BL/6J mice (age 6-8 weeks) were anesthetized and placed into a stereotactic apparatus (David Kopf Instruments, Tujunga, CA). One thousand viable GSC-H cells were injected into the right hemisphere at a position 2 mm lateral to the bregma and 3 mm below the brain surface. After 10 days, animals were exposed to 15 Gy (radiation dose rate, 1.45 Gy/min) with or without prior injection of doranidazole (200 mg/kg, i.p.). Radiation was confined to the brain by protection of the body with a lead shield.
Click to Show/Hide
|
||||
Response regulation | Doranidazole and misonidazole, but not metronidazole, manifested radiation-independent cytotoxicity for hypoxic glioma stem cells (GSCs) that was mediated by ferroptosis induced partially through blockade of mitochondrial complexes I and II and resultant metabolic alterations in oxidative stress responses. | ||||
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target | [29] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | FIN56 | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | |
U118 cells | Astrocytoma | Homo sapiens | CVCL_0633 | ||
In Vivo Model |
1x105 LN229 cells were injected subcutaneously into the right shoulder of 4-week-old nude mice (SLAC laboratory animal Center; Shanghai, China). 2 weeks later, nude mice (n = 10) were divided into two groups, control group and FIN56 treatment group. Subcutaneous tumors were harvested 30 days after treatment. Tumors were fixed and paraffin-embedded for immunohistochemistry.
Click to Show/Hide
|
||||
Response regulation | FIN56 decreased cell viability, inhibited cell proliferation and caused cell cycle arrest on gliomaLN229 and U118 cells. Further study showed that FIN56 induced ferroptosis and induced lysosomal membrane permeabilization in a ferroptosis and transfactor EB dependent manner. Animal study demonstrated that FIN56 inhibited glioma growth and caused ferroptosisin vivo. | ||||
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target | [30] | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Drug | Soyauxinium chloride | Investigative | |||
Pathway Response | Apoptosis | hsa04210 | |||
Ferroptosis | hsa04216 | ||||
Necroptosis | hsa04217 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell necroptosis | |||||
In Vitro Model | AML12 cells | Normal | Mus musculus | CVCL_0140 | |
U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 | ||
Response regulation | The prominent cytotoxic potential of soyauxinium chloride (SCHL) on a panel of 18 human and animal cancer cell lines, including MDR phenotypes. This investigated indoloquinazoline alkaloid induced apoptosis in CCRF-CEM cellsviacaspases activation, MMP alteration and increase of ROS production, and caused ferroptosis and necroptosis in glioblastoma. | ||||
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target | [13] | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Regulator | Neutrophil gelatinase-associated lipocalin (LCN2) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
NF-kappa B signaling pathway | hsa04064 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | Royan N9 cells | Normal | Mus musculus | CVCL_9455 | |
Royan N33 cells | Normal | Mus musculus | CVCL_9417 | ||
T98 cells | Glioblastoma | Homo sapiens | CVCL_B368 | ||
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | ||
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | ||
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | ||
U118 cells | Astrocytoma | Homo sapiens | CVCL_0633 | ||
In Vivo Model |
Four-to five-week-old female BALB/c nude mice were obtained from the Laboratory Animal Center, Southern Medical University. To study the role of IRP1 in TMZ resistance, the mice were randomly divided into four groups (n = 6 per group) (U87TR, U87TR + TMZ, U87TR-lvIRP1, U87TR-lvIRP1 + TMZ). To establish the GBM models, IRP1 overexpress or control U87TR cells (5 x 105 cells per mice in 3 uL PBS) transfected with luciferase lentivirus were injected into the mice brain under the guidance of a stereotactic instrument at coordinates relative to bregma: 2.0 mm posterior, 2.0 mm lateral, and 3.0 mm ventral.
Click to Show/Hide
|
||||
Response regulation | Amplifying IRP1 signals can reverse TMZ resistance and suppress tumor growthin vivo via inhibiting NFKB2 in the noncanonical NF-B signaling pathway. In addition, NFKB2 affected TMZ sensitivity of glioblastoma by modulating the expression of LCN2 and FPN1 in glioblastoma. | ||||
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target | [31] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | TMEM161B-AS1 (IncRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
In this study, 4-week-old specific pathogen free male nude mice were selected and assigned into 4 groups, each of which was respectively injected with U251 cells and U87 cells transfected with NC, hsa-miR-27a-3p angomir, si-TMEM161B-AS1, si-TMEM161B-AS1 + hsa-miR-27a-3p angomir. Each nude mouse was subcutaneously injected with 4 x 105 cells in the right flank area for subcutaneous implantation.
Click to Show/Hide
|
||||
Response regulation | Knockdown of TMEM161B-AS1 down-regulated the expression of FANCD2 and CD44 by sponging hsa-miR-27a-3p, inhibited the proliferation, migration, invasion and promoted apoptosis, ferroptosis of U87 cells and U251 cells. These findings were expected to provide promising therapeutic targets for the treatment of glioma. | ||||
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target | [31] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Fanconi anemia group D2 protein (FANCD2) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
In this study, 4-week-old specific pathogen free male nude mice were selected and assigned into 4 groups, each of which was respectively injected with U251 cells and U87 cells transfected with NC, hsa-miR-27a-3p angomir, si-TMEM161B-AS1, si-TMEM161B-AS1 + hsa-miR-27a-3p angomir. Each nude mouse was subcutaneously injected with 4 x 105 cells in the right flank area for subcutaneous implantation.
Click to Show/Hide
|
||||
Response regulation | Knockdown of TMEM161B-AS1 down-regulated the expression of FANCD2 and CD44 by sponging hsa-miR-27a-3p, inhibited the proliferation, migration, invasion and promoted apoptosis, ferroptosis of glioma U87 cells and U251 cells. These findings were expected to provide promising therapeutic targets for the treatment of glioma. | ||||
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target | [31] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | CD44 antigen (CD44) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
In this study, 4-week-old specific pathogen free male nude mice were selected and assigned into 4 groups, each of which was respectively injected with U251 cells and U87 cells transfected with NC, hsa-miR-27a-3p angomir, si-TMEM161B-AS1, si-TMEM161B-AS1 + hsa-miR-27a-3p angomir. Each nude mouse was subcutaneously injected with 4 x 105 cells in the right flank area for subcutaneous implantation.
Click to Show/Hide
|
||||
Response regulation | Knockdown of TMEM161B-AS1 down-regulated the expression of FANCD2 and CD44 by sponging hsa-miR-27a-3p, inhibited the proliferation, migration, invasion and promoted apoptosis, ferroptosis of U87 cells and U251 cells. These findings were expected to provide promising therapeutic targets for the treatment of glioma. | ||||
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target | [31] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | hsa-miR-27a-3p (miRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Apoptosis | hsa04210 | ||||
Cell Process | Cell ferroptosis | ||||
Cell apoptosis | |||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
In this study, 4-week-old specific pathogen free male nude mice were selected and assigned into 4 groups, each of which was respectively injected with U251 cells and U87 cells transfected with NC, hsa-miR-27a-3p angomir, si-TMEM161B-AS1, si-TMEM161B-AS1 + hsa-miR-27a-3p angomir. Each nude mouse was subcutaneously injected with 4 x 105 cells in the right flank area for subcutaneous implantation.
Click to Show/Hide
|
||||
Response regulation | Knockdown of TMEM161B-AS1 down-regulated the expression of FANCD2 and CD44 by sponging hsa-miR-27a-3p, inhibited the proliferation, migration, invasion and promoted apoptosis, ferroptosis of U87 cells and U251 cells. These findings were expected to provide promising therapeutic targets for the treatment of glioma. | ||||
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target | [32] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Circ-TTBK2 (circRNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell invasion | |||||
In Vitro Model | LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
The stably transfected LN229 cells were set up via the lentivirus-mediation of sh-circ-TTBK2 or sh-NC. Then, the stably transfected LN229 cells (2 x 106 ) were injected into the Balb/c nude mice (4 weeks old, n = 6/group) subcutaneously, which were purchased from Vital River Laboratory Animal Technology (Beijing, China).
Click to Show/Hide
|
||||
Response regulation | Levels of circ-TTBK2 and ITGB8 were upregulated, whereas miR-761 level was low-expressed in glioma tissues and cells. Circ-TTBK2 was a sponge of miR-761 to modulate ITGB8. Additionally, circ-TTBK2 knockdown or miR-761 increase could retard cell proliferation, invasion, and promote ferroptosis in glioma cells. | ||||
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target | [32] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Integrin beta-8 (ITGB8) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell invasion | |||||
In Vitro Model | LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
The stably transfected LN229 cells were set up via the lentivirus-mediation of sh-circ-TTBK2 or sh-NC. Then, the stably transfected LN229 cells (2 x 106 ) were injected into the Balb/c nude mice (4 weeks old, n = 6/group) subcutaneously, which were purchased from Vital River Laboratory Animal Technology (Beijing, China).
Click to Show/Hide
|
||||
Response regulation | Levels of circ-TTBK2 and ITGB8 were upregulated, whereas miR-761 level was low-expressed in glioma tissues and cells. Circ-TTBK2 was a sponge of miR-761 to modulate ITGB8. Additionally, circ-TTBK2 knockdown or miR-761 increase could retard cell proliferation, invasion, and promote ferroptosis in glioma cells. | ||||
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target | [32] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | hsa-miR-761 (miRNA) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell invasion | |||||
In Vitro Model | LN-229 cells | Glioblastoma | Homo sapiens | CVCL_0393 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
The stably transfected LN229 cells were set up via the lentivirus-mediation of sh-circ-TTBK2 or sh-NC. Then, the stably transfected LN229 cells (2 x 106 ) were injected into the Balb/c nude mice (4 weeks old, n = 6/group) subcutaneously, which were purchased from Vital River Laboratory Animal Technology (Beijing, China).
Click to Show/Hide
|
||||
Response regulation | Levels of circ-TTBK2 and ITGB8 were upregulated, whereas miR-761 level was low-expressed in glioma tissues and cells. Circ-TTBK2 was a sponge of miR-761 to modulate ITGB8. Additionally, circ-TTBK2 knockdown or miR-761 increase could retard cell proliferation, invasion, and promote ferroptosis in glioma cells. | ||||
Experiment 16 Reporting the Ferroptosis-centered Disease Response by This Target | [33] | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Multidrug resistance-associated protein 1 (ABCC1) | Driver | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell invasion | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
T98G cells | Glioblastoma | Homo sapiens | CVCL_0556 | ||
Response regulation | NRF2 positively correlates with ABCC1 expression in tumor tissues of glioma patients, which can be associated with tumor aggressiveness, drug resistance, and poor overall survival. | ||||
Long-chain-fatty-acid--CoA ligase 4 (ACSL4)
In total 2 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [16] | |||
Target for Ferroptosis | Driver | |||
Responsed Disease | Glioma [ICD-11: 2A00] | |||
Responsed Drug | Dihydrotanshinone I | Investigative | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model | HEB (Human glial cells) | |||
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
Response regulation | Dihydrotanshinone I (DHI) inhibited the proliferation of human glioma cells. Following treatment of the U251 and U87 cells with DHI, changes in the expression levels of ferroptosis-associated proteins were observed; the expression level of GPX4 decreased and that of ACSL-4 increased. DHI also increased the levels of LDH and MDA in the human glioma cells and reduced the GSH/GSSG ratio. | |||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [22] | |||
Target for Ferroptosis | Driver | |||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | |||
Responsed Regulator | hsa-miR-670-3p (miRNA) | Suppressor | ||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 |
A-172 cells | Glioblastoma | Homo sapiens | CVCL_0131 | |
SVG p12 cells | Normal | Homo sapiens | CVCL_3797 | |
hMGCs (Human normal brain astroglia cells) | ||||
Response regulation | miR-670-3p level was elevated in human glioblastoma, but decreased upon ferroptotic stimulation. Mechanistically, ACSL4 was required for the regulation on ferroptosis and growth of glioblastoma cells by miR-670-3p. Moreover, U87MG and A172 cells treated with miR-670-3p inhibitor showed an increased chemosensitivity to TMZ. | |||
Hydroperoxide isomerase ALOXE3 (ALOXE3)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [23] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Regulator | hsa-mir-18a (Precursor RNA) | Suppressor | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
The 8-week-old male nude mice were randomly divided into two groups (6 mice per group). The investigators were not blinded to the experimental groups. Orthotopic implantation of GBM cells into the hippocampus of nude mice was performed as we previously described (n = 6). Mice were intraperitoneally injected with luciferin (150 mg/kg; catalog #P1043; Promega) and subjected to IVIS Spectrum in vivo imaging system (PerkinElmer) to determine the intracranial tumor size.
Click to Show/Hide
|
||||
Response regulation | MiR-18a/ALOXE3 axis exerts tumor promoting functions by regulating ferroptosis and migration of glioblastoma cells. Mechanistically, miR-18a directly targeted ALOXE3 and suppressed its expression and functions in GBM cells. | ||||
Heme oxygenase 1 (HMOX1)
In total 3 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [24] | ||||
Target for Ferroptosis | Driver/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Siramesine | Terminated | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
Response regulation | Lapatinib and siramesine was the most effective tyrosine kinase inhibitor and lysosome disruptor drug combination in inducing synergistic cell death in A549 and U87 cells. This cell death was through ferroptosis mediated by ROS and reduced expression of HO-1 in glioma cells. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [9] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Dihydroartemisinin | Investigative | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
All BALB/C nude mice were purchased from Huafukang Biotechnology (Beijing, China). These mice were 5 weeks old and weighed 14-16 g. We established subcutaneous tumour-forming mouse model by injecting 5 x 106 U87 cells into the lateral abdomen of BALB/C nude mice. Animals were then treated with DHA solvent (50 mg/kg) by intragastric administration once a day for 26 days.
Click to Show/Hide
|
||||
Response regulation | Dihydroartemisinin could promote ferroptosis in glioma cells. Low expression of GPX4 and high expression of HMOX1 were identified in DHA treated glioma cells. MAZ was further identified as the direct target of long noncoding RNA (lncRNA) TUG1 through luciferase assay. Downregulated expression of TUG1 and upregulated expression of MAZ were identified in DHA treated glioma cells. TUG1 overexpression or inhibition of FTH1 expression could enhance the antiglioma effect of DHA in vitro and in vivo, providing a promising strategy to enhance the antitumor effect of DHA in glioma. | ||||
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target | [24] | ||||
Target for Ferroptosis | Driver/Suppressor | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Lapatinib | Investigative | |||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
A-549 cells | Lung adenocarcinoma | Homo sapiens | CVCL_0023 | ||
Response regulation | Lapatinib and siramesine was the most effective tyrosine kinase inhibitor and lysosome disruptor drug combination in inducing synergistic cell death in A549 and U87 cells. This cell death was through ferroptosis mediated by ROS and reduced expression of HO-1 in glioma cells. | ||||
GTP cyclohydrolase 1 (GCH1)
In total 2 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [25] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Regulator | Paired mesoderm homeobox protein 2 (PRRX2) | Suppressor | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | GSC51 (WHO grade IV specimens) | ||||
GSC52 (WHO grade IV specimens) | |||||
GSC53 (WHO grade IV specimens) | |||||
GSC55 (WHO grade IV specimens) | |||||
GSC56 (WHO grade IV specimens) | |||||
GSC58 (WHO grade IV specimens) | |||||
In Vivo Model |
Five-week-old female BALB/c nude mice were purchased from Shanghai Jihui Laboratory Animal Care Co., Ltd (Shanghai, China). All mice were bred in the Laboratory Animal Center of Shanghai Tenth Peoples Hospital under specific pathogen-free conditions. The animal experiments were performed by the Animal Care Committee of Shanghai Tenth Peoples Hospital. Briefly, each group contains five mice, and 5 x 104 GSCs were injected orthotopically into the mouse brain at 2 mm lateral and 2 mm anterior to the bregma with a stereotaxic apparatus. Then the survival time of each mouse was calculated, and the tumor volume was calculated according to the formula: V = (D x d2) / 2, where D represents the longest diameter and d represents the shortest diameter.
Click to Show/Hide
|
||||
Response regulation | CircLRFN5 can be used as a potential Glioblastoma biomarker and become a target for molecular therapies or ferroptosis-dependent therapy in GBM. Mechanistically, circLRFN5 binds to PRRX2 protein and promotes its degradation via a ubiquitin-mediated proteasomal pathway. PRRX2 can transcriptionally upregulate GCH1 expression in GSCs, which is a ferroptosis suppressor via generating the antioxidant tetrahydrobiopterin (BH4). | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [25] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Responsed Regulator | CircLRFN5 (circRNA) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
In Vitro Model | GSC51 (WHO grade IV specimens) | ||||
GSC52 (WHO grade IV specimens) | |||||
GSC53 (WHO grade IV specimens) | |||||
GSC55 (WHO grade IV specimens) | |||||
GSC56 (WHO grade IV specimens) | |||||
GSC58 (WHO grade IV specimens) | |||||
In Vivo Model |
Five-week-old female BALB/c nude mice were purchased from Shanghai Jihui Laboratory Animal Care Co., Ltd (Shanghai, China). All mice were bred in the Laboratory Animal Center of Shanghai Tenth Peoples Hospital under specific pathogen-free conditions. The animal experiments were performed by the Animal Care Committee of Shanghai Tenth Peoples Hospital. Briefly, each group contains five mice, and 5 x 104 GSCs were injected orthotopically into the mouse brain at 2 mm lateral and 2 mm anterior to the bregma with a stereotaxic apparatus. Then the survival time of each mouse was calculated, and the tumor volume was calculated according to the formula: V = (D x d2) / 2, where D represents the longest diameter and d represents the shortest diameter.
Click to Show/Hide
|
||||
Response regulation | CircLRFN5 can be used as a potential Glioblastoma biomarker and become a target for molecular therapies or ferroptosis-dependent therapy in GBM. Mechanistically, circLRFN5 binds to PRRX2 protein and promotes its degradation via a ubiquitin-mediated proteasomal pathway. PRRX2 can transcriptionally upregulate GCH1 expression in GSCs, which is a ferroptosis suppressor via generating the antioxidant tetrahydrobiopterin (BH4). | ||||
Glutaminase liver isoform, mitochondrial (GLS2)
In total 2 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [26] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | Double-stranded RNA-specific adenosine deaminase (ADAR) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U251-TR3 cells | Astrocytoma | Homo sapiens | CVCL_1G29 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Nude mice (four-week-old, female, 3/group) were purchased from Model Animal Research Center of Nanjing University. 2.5 x 105 luciferase labeled pcDNA3.1-ATXN8OS/GLS2 were transfected into U251 cells. After 48 h, transfected cells were injected stereotactically into thebrainof nude mice. In the first week after operation, TMZ was given to treat nude mice by oral gavage (66 mg/kg per day for 5 days), with vehicle as control. Bioluminescence imaging (IVIS Spectrum; PerkinElmer, USA) was applied for confirming the formation of intracranial tumor. 28 days late, all the mice were sacrificed. The resected tumors were measure at Lipid ROS and intracellular iron level. The whole experiments were approved by ethic committee of Shanxi Provincial Peoples Hospital.
Click to Show/Hide
|
||||
Response regulation | Rescue experiments indicated that ATXN8OS modulated TMZ-resistance of glioma through GLS2. In conclusion, ATXN8OS mediated ferroptosis and regulated the TMZ-resistance of glioma via ADAR/GLS2 pathway. | ||||
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target | [26] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Regulator | ATXN8OS (IncRNA) | Driver | |||
Pathway Response | Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | ||||
In Vitro Model | U251-TR3 cells | Astrocytoma | Homo sapiens | CVCL_1G29 | |
U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | ||
In Vivo Model |
Nude mice (four-week-old, female, 3/group) were purchased from Model Animal Research Center of Nanjing University. 2.5 x 105 luciferase labeled pcDNA3.1-ATXN8OS/GLS2 were transfected into U251 cells. After 48 h, transfected cells were injected stereotactically into thebrainof nude mice. In the first week after operation, TMZ was given to treat nude mice by oral gavage (66 mg/kg per day for 5 days), with vehicle as control. Bioluminescence imaging (IVIS Spectrum; PerkinElmer, USA) was applied for confirming the formation of intracranial tumor. 28 days late, all the mice were sacrificed. The resected tumors were measure at Lipid ROS and intracellular iron level. The whole experiments were approved by ethic committee of Shanxi Provincial Peoples Hospital.
Click to Show/Hide
|
||||
Response regulation | Rescue experiments indicated that ATXN8OS modulated TMZ-resistance of glioma through GLS2. In conclusion, ATXN8OS mediated ferroptosis and regulated the TMZ-resistance of glioma via ADAR/GLS2 pathway. | ||||
Beclin-1 (BECN1)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [8] | ||||
Target for Ferroptosis | Driver | ||||
Responsed Disease | Glioma [ICD-11: 2A00] | ||||
Responsed Drug | Amentoflavone | Investigative | |||
Pathway Response | mTOR signaling pathway | hsa04150 | |||
Fatty acid metabolism | hsa01212 | ||||
Autophagy | hsa04140 | ||||
Cell Process | Cell ferroptosis | ||||
Cell autophagy | |||||
Cell proliferation | |||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 | |
U-373MG cells | Astrocytoma | Homo sapiens | CVCL_2219 | ||
In Vivo Model |
1 x 107 U251 cells were subcutaneously injected into the right back of the four-week-old BALB/c nude mice. After the tumor grown to 100 mm3, mice were randomly divided into 3 groups: mice in control group receivedintraperitoneal injectionof saline, while mice in AF treatment group received intraperitoneal injection at dosages of 40 mg/kg/day or 80 mg/kg/day, respectively.
Click to Show/Hide
|
||||
Response regulation | Amentoflavone (AF) triggered ferroptosis in autophagy-dependent manner. Furthermore, the expression of LC3B, Beclin1, ATG5, ATG7 were increased, and the expression of FTH were decreased by AF in a dose-dependent manner in vivo. AF has the potential to be considered as a novel treatment agent in glioma. | ||||
Transferrin receptor protein 2 (TFR2)
In total 1 item(s) under this target | ||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [34] | |||
Target for Ferroptosis | Driver | |||
Responsed Disease | Glioma [ICD-11: 2A00] | |||
Pathway Response | Fatty acid metabolism | hsa01212 | ||
Ferroptosis | hsa04216 | |||
Cell Process | Cell ferroptosis | |||
Cell proliferation | ||||
In Vitro Model | U-251MG cells | Astrocytoma | Homo sapiens | CVCL_0021 |
U87 MG-Red-Fluc cells | Glioblastoma | Homo sapiens | CVCL_5J12 | |
Response regulation | Overexpression of TFR2 promoted the production of reactive oxygen species and lipid peroxidation in glioma cells, thereby further promoting ferroptosis. In conclusion, TFR2 induced ferroptosis and enhanced TMZ sensitivity in gliomas. | |||
Cytochrome b-245 heavy chain (CYBB)
In total 1 item(s) under this target | |||||
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target | [35] | ||||
Target for Ferroptosis | Suppressor | ||||
Responsed Disease | Glioblastoma [ICD-11: 2A00] | ||||
Pathway Response | Fatty acid metabolism | hsa01212 | |||
Ferroptosis | hsa04216 | ||||
Cell Process | Cell ferroptosis | ||||
Cell proliferation | |||||
Cell migration | |||||
Cell invasion | |||||
In Vitro Model | U-87MG cells | Glioblastoma | Homo sapiens | CVCL_GP63 | |
In Vivo Model |
A total of four groups were formed by randomly dividing the mice: control group consisting of shScramble xenografts treated with vehicle (n = 5), shScramble xenografts treated with IKE (n = 5), shSOD2 xenografts treated with vehicle (n = 5), and shSOD2 xenografts treated with IKE (n = 5). Drug treatment was started after the tumor volume reached approximately 100 mm3 or 10 days after the injection of the tumor xenograft. Depending on the treatment group, vehicles (5 mg/kg/day) or IKE (25 mg/kg/day) were administered intraperitoneally. Treatment was administered intraperitoneally for 3 weeks, and tumor growth was observed for 6 weeks after treatment began.
Click to Show/Hide
|
||||
Response regulation | CYBB captured ferroptosis resilience in mesenchymal glioblastoma multiforme (GBM). The downstream compensatory activity of CYBB via the Nrf2/SOD2 axis is exploitable through erastin-induced ferroptosis to overcome TMZ resistance. | ||||
References