General Information of the Disease (ID: DIS00028)
Name
Colorectal cancer
ICD
ICD-11: 2B91
Full List of Target(s) of This Ferroptosis-centered Disease
Phospholipid hydroperoxide glutathione peroxidase (GPX4)
In total 18 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Curcumin Investigative
Responsed Regulator Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform (PIK3CA) Suppressor
Pathway Response PI3K-Akt signaling pathway hsa04151
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HCT-8 cells Ileocecal adenocarcinoma Homo sapiens CVCL_2478
Response regulation Treating HCT-8 cells with curcumin significantly downregulated GSH, SLC7A11, and GPX4, while significantly increasing levels of iron, MDA, and ROS. Curcumin triggers ferroptosis and suppresses proliferation of colorectal cancer cells by inhibiting the PI3K/Akt/mTOR signaling pathway.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Curcumin Investigative
Responsed Regulator Serine/threonine-protein kinase mTOR (MTOR) Suppressor
Pathway Response PI3K-Akt signaling pathway hsa04151
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HCT-8 cells Ileocecal adenocarcinoma Homo sapiens CVCL_2478
Response regulation Treating HCT-8 cells with curcumin significantly downregulated GSH, SLC7A11, and GPX4, while significantly increasing levels of iron, MDA, and ROS. Curcumin triggers ferroptosis and suppresses proliferation of colorectal cancer cells by inhibiting the PI3K/Akt/ mTOR signaling pathway.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Curcumin Investigative
Responsed Regulator RAC-alpha serine/threonine-protein kinase (AKT1) Suppressor
Pathway Response PI3K-Akt signaling pathway hsa04151
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HCT-8 cells Ileocecal adenocarcinoma Homo sapiens CVCL_2478
Response regulation Treating HCT-8 cells with curcumin significantly downregulated GSH, SLC7A11, and GPX4, while significantly increasing levels of iron, MDA, and ROS. Curcumin triggers ferroptosis and suppresses proliferation of colorectal cancer cells by inhibiting the PI3K/ Akt/mTOR signaling pathway.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Curcumin Investigative
Responsed Regulator Serine/threonine-protein kinase mTOR (MTOR) Suppressor
Pathway Response PI3K-Akt signaling pathway hsa04151
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HCT-8 cells Ileocecal adenocarcinoma Homo sapiens CVCL_2478
Response regulation Treating HCT-8 cells with curcumin significantly downregulated GSH, SLC7A11, and GPX4, while significantly increasing levels of iron, MDA, and ROS. Curcumin triggers ferroptosis and suppresses proliferation of colorectal cancer cells by inhibiting the PI3K/Akt/mTOR signaling pathway.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [1]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Curcumin Investigative
Responsed Regulator RAC-alpha serine/threonine-protein kinase (AKT1) Suppressor
Pathway Response PI3K-Akt signaling pathway hsa04151
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HCT-8 cells Ileocecal adenocarcinoma Homo sapiens CVCL_2478
Response regulation Treating HCT-8 cells with curcumin significantly downregulated GSH, SLC7A11, and GPX4, while significantly increasing levels of iron, MDA, and ROS. Curcumin triggers ferroptosis and suppresses proliferation of colorectal cancer cells by inhibiting the PI3K/Akt/mTOR signaling pathway.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Andrographis Approved
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
Briefly, surgically resected tumors were maintained in DMEM-F12 (Gibco) supplemented with 1% HEPES (Sigma-Aldrich), 1% L-glutamine (Gibco), 10% FBS (Gibco), 2% penicillin/streptomycin (Sigma-Aldrich), and 10 uM Y-27632 (R&D Systems). Tumors were digested with collagenase solution (5 mL of the above medium with 75 uL collagenase, 124 ug/mL dispase type II, and 0.2% Primocen) for 30 min and then filtered through a 70 um filter (Corning). An organoid pellet was obtained by centrifugation (200x g for 10 min). Organoids were suspended in Matrigel (Corning, Tehama County, CA) with IntestiCult Organoid Growth Medium (#06010, STEMCELL Technologies) and seeded in 12-well plates. Approximately 750 uL of IntestiCult Organoid Growth Medium was added to each well. Organoids were divided into five groups of control, curcumin (3.0 ug/mL), andrographis (30.0 ug/mL), their combination (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL), and their combination plus ferrostatin-1 (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL; ferrostatin-1; 20 uM). Following forty-eight hours of treatment, the numbers of organoids (<100 um) and their mean sizes were examined using Image J software.

    Click to Show/Hide
Response regulation In conclusion, our study revealed that combined treatment with curcumin and andrographis exhibited anti-tumorigenic effects in colorectal cancer cells through activation of ferroptosis and by dual suppression of GPX-4 and FSP-1, which have significant potential implications for the adjunctive treatment of CRC patients. This combination treatment resulted in cancer cell death via both forms of cell death: apoptosis and ferroptosis.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [13]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug RSL3 Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
HT29 cells Colon cancer Mus musculus CVCL_A8EZ
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
Response regulation RSL3 triggered ferroptotic cell death by promoting the accumulation of cellular ROS and increasing the cellular LIP level. Mechanismly, we found transferrin expression were elevated in colorectal cancer cells treated with RSL3 accompanied by a decrease in the expression of GPX4, indicating an iron-dependent cell death.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Curcumin Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
Briefly, surgically resected tumors were maintained in DMEM-F12 (Gibco) supplemented with 1% HEPES (Sigma-Aldrich), 1% L-glutamine (Gibco), 10% FBS (Gibco), 2% penicillin/streptomycin (Sigma-Aldrich), and 10 uM Y-27632 (R&D Systems). Tumors were digested with collagenase solution (5 mL of the above medium with 75 uL collagenase, 124 ug/mL dispase type II, and 0.2% Primocen) for 30 min and then filtered through a 70 um filter (Corning). An organoid pellet was obtained by centrifugation (200x g for 10 min). Organoids were suspended in Matrigel (Corning, Tehama County, CA) with IntestiCult Organoid Growth Medium (#06010, STEMCELL Technologies) and seeded in 12-well plates. Approximately 750 uL of IntestiCult Organoid Growth Medium was added to each well. Organoids were divided into five groups of control, curcumin (3.0 ug/mL), andrographis (30.0 ug/mL), their combination (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL), and their combination plus ferrostatin-1 (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL; ferrostatin-1; 20 uM). Following forty-eight hours of treatment, the numbers of organoids (<100 um) and their mean sizes were examined using Image J software.

    Click to Show/Hide
Response regulation In conclusion, our study revealed that combined treatment with curcumin and andrographis exhibited anti-tumorigenic effects in colorectal cancer cells through activation of ferroptosis and by dual suppression of GPX-4 and FSP-1, which have significant potential implications for the adjunctive treatment of CRC patients. This combination treatment resulted in cancer cell death via both forms of cell death: apoptosis and ferroptosis.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [14]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator hsa-mir-539 (Precursor RNA) Driver
Pathway Response Ferroptosis hsa04216
MAPK signaling pathway hsa04010
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
HEK-293T cells Normal Homo sapiens CVCL_0063
In Vivo Model
Six 4-week-old male BALB/c nude mice were ordered from the Shanghai Laboratory Animal Center (Shanghai SLAC Laboratory Animal Co., Ltd., China). A total of 5 x 106 TIPE+/+ SW480 cells were suspended in 100 uL of PBS and subcutaneously injected into the right axilla flank of each nude mouse, and the same amount of vector SW480 cells was into the left. At 2 weeks after inoculation, the xenograft tumor size was measured using Vernier calipers every 2 days.

    Click to Show/Hide
Response regulation MiR-539 can bind to and regulate the expression of TIPE, and miR-539 activates SAPK/JNK to downregulate the expression of glutathione peroxidase 4 (GPX4) and promote ferroptosis. In addition, SAPK/JNK is the upstream molecule of p53. MiR-539 is a new therapeutic target for colorectal cancer (CRC) patients.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [14]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Cellular tumor antigen p53 (TP53) Driver
Pathway Response Ferroptosis hsa04216
MAPK signaling pathway hsa04010
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
HEK-293T cells Normal Homo sapiens CVCL_0063
In Vivo Model
Six 4-week-old male BALB/c nude mice were ordered from the Shanghai Laboratory Animal Center (Shanghai SLAC Laboratory Animal Co., Ltd., China). A total of 5 x 106 TIPE+/+ SW480 cells were suspended in 100 uL of PBS and subcutaneously injected into the right axilla flank of each nude mouse, and the same amount of vector SW480 cells was into the left. At 2 weeks after inoculation, the xenograft tumor size was measured using Vernier calipers every 2 days.

    Click to Show/Hide
Response regulation MiR-539 can bind to and regulate the expression of TIPE, and miR-539 activates SAPK/JNK to downregulate the expression of glutathione peroxidase 4 (GPX4) and promote ferroptosis. In addition, SAPK/JNK is the upstream molecule of p53. MiR-539 is a new therapeutic target for colorectal cancer (CRC) patients.
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target [15]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Metalloproteinase inhibitor 1 (TIMP1) Suppressor
Pathway Response Ferroptosis hsa04216
PI3K-Akt signaling pathway hsa04151
Cell Process Cell ferroptosis
In Vitro Model HCT-8 cells Ileocecal adenocarcinoma Homo sapiens CVCL_2478
Response regulation TIMP1 depletion in colorectal cancer cells enhances sorafenib-triggered ferroptosis by reducing PI3K/Akt axis signal transduction. TIMP1 knockdown repressed the activation of the PI3K/Akt pathway and reduced levels of glutathione peroxidase 4 (GPX4), enhancing sorafenib-induced ferroptosis.
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target [16]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Endoplasmic reticulum chaperone BiP (HSPA5) Suppressor
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
Male BALB/c nude mice (4-6 weeks) were purchased from the Air Force Medical University Laboratory Animal Center. The mice were kept in the SPF environment and had free access to food and water. 3 x 106 SW480 cells were injected subcutaneously into nude mice (n = 4 or 5). Erastin was dissolved in 5% DMSO/corn oil and intraperitoneally injected into nude mice at a dose of 15 mg/kg three times. Three weeks later, mice were anesthetized by intraperitoneal injection of 10% chloral hydrate (35 mg/kg). When mice were successfully anesthetized five minutes later, mice were sacrificed and the tumors were resected and weighed. The tumors were divided into two parts. One sample was lysed and used for protein analysis. The other part was used to test for Ki67 expression.

    Click to Show/Hide
Response regulation HSPA5 restrained ferroptosis to promote colorectal cancer development by maintaining GPX4 stability. HSPA5 was demonstrated to play a diagnostic role and correlated to the immune microenvironment in CRC patients.
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target [17]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Nitric oxide synthase, inducible (NOS2) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
NF-kappa B signaling pathway hsa04064
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
In Vivo Model
All nude mice were purchased from Guangdong Medical Laboratory Animal Center. NOS2-overexpressing and control cell lines were transplanted subcutaneously into the bilateral flanks, and appropriate care was given to these animals.

    Click to Show/Hide
Response regulation Ferroptosis-related genes (FRGs) have potential prognostic value in colorectal cancer patients and that NOS2 suppresses tumor progression, providing a novel therapeutic target for CRC treatment based on ferroptosis. And NOS2 overexpression in CACO2 cells decreased the expression of GPX4.
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target [18]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Short/branched chain specific acyl-CoA dehydrogenase, mitochondrial (ACADSB) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
Cell migration
Cell invasion
In Vitro Model SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
Response regulation Overexpression of ACADSB inhibits colorectal cancer cell migration, invasion, and proliferation, while ACADSB knockdown has the opposite effect. More importantly, ACADSB negatively regulates expression of glutathione reductase and glutathione peroxidase 4 (GPX4), the two main enzymes responsible for clearing glutathione (GSH) in CRC cells.
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target [19]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Serine/arginine-rich splicing factor 9 (SRSF9) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
LOVO cells were stably transfected with SRSF9-shRNA1 or NC-shRNA. Caco-2 cells were stably transfected with SRSF9-OE or empty vector. Then the transfected CRC cells (2*105 cells/100 uL) were injected into the right armpit of mouse 6-8-week-old male athymic nude mice. When the tumors reached 50 mm3 at day 7, erastin (40 mg/kg) was administrated to mice by intraperitoneal injection twice every other day.

    Click to Show/Hide
Response regulation SRSF9's regulation of GPX4 as an essential mechanism driving colorectal cancer (CRC) tumorigenesis and resistance of erastin-induced ferroptosis. This molecular mechanism may provide a novel method for improving the sensitivity of CRC to erastin.
Experiment 16 Reporting the Ferroptosis-centered Disease Response by This Target [20]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator WD repeat domain phosphoinositide-interacting protein 2 (WIPI2) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
HT29 cells Colon cancer Mus musculus CVCL_A8EZ
Response regulation The expression level of ACSL4 decreased and that of GPX4 increased when WIPI2 was knocked down, suggesting that WIPI2 can potentially positively regulate colorectal cancer ferroptosis.
Experiment 17 Reporting the Ferroptosis-centered Disease Response by This Target [21]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator hsa-miR-15a-3p (miRNA) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
HT29 cells Colon cancer Mus musculus CVCL_A8EZ
KM12 cells Colon carcinoma Homo sapiens CVCL_1331
NCM460 cells Normal Homo sapiens CVCL_0460
In Vivo Model
We obtained the 6-week-old nude mice (BALB/c) from Beijing HFK Bioscience Co., Ltd. and randomly divided into eight groups (n = 5/group): (a) NC + dimethyl sulfoxide (DMSO) group; (b) NC + erastin group; (c) mimic + DMSO group; (d) mimic + erastin group; (e) iNC + DMSO group; (f) iNC + erastin group; (g) inhibitor + DMSO group; (h) inhibitor + erastin group. Control and transfected cells (7 x 106) were subcutaneously injected into the nude mice. After the tumor sizes reached roughly >50 mm3, mice in Groups B, D, F, and H were treated with 15 mg/kg erastin twice every day for about 20 days. Meanwhile, mice in Groups A, C, E, and G were treated with an equal volume DMSO. Besides, tumor formation and mass were monitored and the size of tumor was counted using the formula V = 0.5 x L x W2 (L, length and W, width). At last, after killing the nude mice and then we isolated the mice tumor tissues.

    Click to Show/Hide
Response regulation Overexpression of miR-15a-3p repressed GPX4 through binding to the 3'-untranslated region of GPX4, resulting in increased reactive oxygen species level, intracellular Fe2+ level, and malondialdehyde accumulation in vitro and in vivo. MiR-15a-3p suppressed colorectal cancer cell growth and enhanced cell ferroptosis by inactivating GPX4.
Experiment 18 Reporting the Ferroptosis-centered Disease Response by This Target [22]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator LINC00239 (IncRNA) Suppressor
Pathway Response Pathways in cancer hsa05200
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model RKO cells Colon carcinoma Homo sapiens CVCL_0504
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
FHC cells Normal Homo sapiens CVCL_3688
In Vivo Model
To clarify the role of LINC00239 in vivo, we used 4-week-old male BALB/c nude mice provided by the Experimental Animal Center of the Air Force Military Medical University. HCT116 or SW620 cells (1 x 107 cells) were injected subcutaneously into the right flanks of these mice to establish a CRC xenograft model. One week after the injection of cells, the volume of xenografts was continuously monitored (once a week). Four weeks later, the xenografts were removed, and the weights were measured.

    Click to Show/Hide
Response regulation LINC00239 plays a novel and indispensable role in ferroptosis by nucleotides 1-315 of LINC00239 to interact with the Kelch domain (Nrf2-binding site) of Keap1, inhibiting Nrf2 ubiquitination and increasing Nrf2 protein stability. And LINC00239 expression has a positive correlation with Nrf2 and GPX4 expression in colorectal cancer tissues. LINC00239 inhibition in combination with ferroptosis induction might be a promising therapeutic strategy for CRC patients.
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 Colorectal cancer [ICD-11: 2B91]
Responsed Drug Cetuximab Approved
Responsed Regulator Mitogen-activated protein kinase 14 (MAPK14) Driver
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
The DLD-1 cell suspension (4 x 106 cells/200 ul) was injected subcutaneously into the right dorsal flank of 5-week-old male BALB/c nude mice (Charles River, China). The mice were randomly divided into four groups (5 mice/group): 1) the control group, 2) the RSL3 group, 3) the cetuximab group, and 4) the RSL3 + cetuximab group. Both RSL3 (5 mg/kg) and cetuximab (13 mg/kg) were administered by intraperitoneal injection in a volume of 100 ul once per day. The tumour volume was calculated as 0.5 x length x width2. After 17 days of treatment, the mice were sacrificed, and the tumours were removed. Then, tumour tissue obtained from the different treated groups was subjected to western blotting and immunohistochemical experiments.

    Click to Show/Hide
Response regulation Our work reveals that cetuximab enhances the cytotoxic effect of RSL3 on KRAS mutant Colorectal cancer (CRC) cells and that cetuximab enhances RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 axis through the activation of p38 MAPK.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [3]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Ibrutinib Investigative
Responsed Regulator Tyrosine-protein kinase BTK (BTK) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
LS513 cells Cecum adenocarcinoma Homo sapiens CVCL_1386
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
SW1116 cells Colon adenocarcinoma Homo sapiens CVCL_0544
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
HT-29 cells Colon adenocarcinoma Homo sapiens CVCL_0320
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
In Vivo Model
Sixty mice were randomly divided into six groups, (1) the CRC model group (model), (2) mice with RSL3 treatment, (3) mice with Erastin treatment, (4) mice with Ibrutinib treatment, (5) mice with RSL3 and Ibrutinib treatment, and (6) Erastin and Ibrutinib group. Murine subcutaneous tumor model and xenograft tumor mouse model were established and please refer to supplemental method for details. For CRC model group, the mice were treated with PBS for two weeks. For RSL3 group, the mice were intraperitoneal injected with RSL3 (5 mg/kg daily) for two weeks. For Erastin group, the mice were intraperitoneal injected with Erastin (30 mg/kg, twice every other day) for two weeks. For Ibrutinib treatment group, mice were administered in drinking water at a concentration of 0.16 mg/ml for two weeks. Mice were also treated in combination with RSL and Ibrutinib or Erastin and Ibrutinib.

    Click to Show/Hide
Response regulation Ibrutinib inhibited BTK, which prevented Nrf2 translocating to cell nucleus and the activation of the Nrf2 dependent antioxidant genes during oxidative stress conditions and eventually enhanced the sensitivity of Colorectal cancer (CRC) cells to ferroptosis.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [23]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Oxaliplatin Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HT29 cells Colon cancer Mus musculus CVCL_A8EZ
Response regulation Oxaliplatin promoted ferroptosis and oxidative stress in colorectal cancer cells by inhibiting the Nrf2 signaling pathway. Treatment with oxaliplatin enhanced the effects of erastin on CRC cells by promoting ferroptosis and oxidative stress and inhibiting cell viability.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Propofol Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HT29 cells Colon cancer Mus musculus CVCL_A8EZ
CT26 cells Colon adenocarcinoma Mus musculus CVCL_7254
In Vivo Model
CT26 (1 x 105 cells/100 uL) were injected into thetail veinof male BALB/c mice. Then the mice were randomly divided into saline, vehicle, propofol, and sevoflurane groups (n = 5 per group). Saline, fat emulsion (as vehicle control of propofol), and propofol (200 mg/kg) were intraperitoneally injected, while sevoflurane (1.8-2.0%) was administered by inhalation for 2 h. In another set of experiments, coloncancer cells (CT26 and HT29) were pretreated with two doses of propofol (5 ug/mL, 10 ug/mL) or fat emulsion (as vehicle control of propofol) in a cell culture medium for 2 h. After washing with phosphate-buffered saline (PBS), the cells were harvested,counted on a hemacytometer and prepared. Cells (CT26: 1 x 105 cells/100 uL, HT29: 1 x 106 cells/100 uL) were finnally injected into mice through the tail vein.Lung metastasiswas detected via hematoxylin and eosin staining (HE) or ex vivo bioluminescence imaging.

    Click to Show/Hide
Response regulation Further studies showed that propofol treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target genes, including HO-1, NQO1, and SLC7A11. Collectively, we demonstrated the risk of a specific type of anesthetic, propofol, in promoting colorectal cancer cell metastasis through Nrf2-mediated ferroptosis inhibition.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [25]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug tagitinin C Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Response regulation Tagitinin C induces ferroptosis in colorectal cancer cells and has synergistic effect together with erastin. Mechanistically, tagitinin C induces ferroptosis through ER stress-mediated activation of PERK-Nrf2-HO-1 signaling pathway.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [26]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Kinesin-like protein KIF20A (KIF20A) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
NCI-H716 cells Cecum adenocarcinoma Homo sapiens CVCL_1581
In Vivo Model
HCT116-Or or H716 cells (7*106) were suspended into 200 ul matrigel (BD Bioscience), and injected into the subcutaneous tissues of mouse right lower limbs. Animals were grouped randomly (eight mouse per group) and medication was initiated when the average xenograft size was over 100 mm3: Oxaliplatin was given alone weekly through intraperitoneal injection (5 mg/kg), or in combination with liproxstatin-1 through intraperitoneal injection for twice a week (125 mg/kg) and RSL3 through intra-tumor injection (100 mg/kg, in order to achieve better local concentration and reduce the probable systemic toxicity of RSL3) weekly.

    Click to Show/Hide
Response regulation KIF20A was highly expressed in the oxaliplatin-resistant cell lines and was strongly correlated with survival among colorectal cancer patients. Silencing KIF20A enhanced cellular sensitivity to oxaliplatin both in vivo and in vitro, and silencing KIF20A also suppressed NUAK1 activation. Moreover, silencing NUAK1 up-regulated the expression of PP1, down-regulated the phosphorylation of downstream GSK3Ser9, suppressed the nuclear import of Nrf2, inhibited the expression of a ferroptosis key negative regulatory protein (GPX4), and blocked cellular resistance.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [22]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Kelch-like ECH-associated protein 1 (KEAP1) Driver
Pathway Response Pathways in cancer hsa05200
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model RKO cells Colon carcinoma Homo sapiens CVCL_0504
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
FHC cells Normal Homo sapiens CVCL_3688
In Vivo Model
To clarify the role of LINC00239 in vivo, we used 4-week-old male BALB/c nude mice provided by the Experimental Animal Center of the Air Force Military Medical University. HCT116 or SW620 cells (1 x 107 cells) were injected subcutaneously into the right flanks of these mice to establish a CRC xenograft model. One week after the injection of cells, the volume of xenografts was continuously monitored (once a week). Four weeks later, the xenografts were removed, and the weights were measured.

    Click to Show/Hide
Response regulation LINC00239 plays a novel and indispensable role in ferroptosis by nucleotides 1-315 of LINC00239 to interact with the Kelch domain (Nrf2-binding site) of Keap1, inhibiting Nrf2 ubiquitination and increasing Nrf2 protein stability. And LINC00239 expression has a positive correlation with Nrf2 and GPX4 expression in colorectal cancer tissues. LINC00239 inhibition in combination with ferroptosis induction might be a promising therapeutic strategy for CRC patients.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [26]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator NUAK family SNF1-like kinase 1 (NUAK1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
NCI-H716 cells Cecum adenocarcinoma Homo sapiens CVCL_1581
In Vivo Model
HCT116-Or or H716 cells (7*106) were suspended into 200 ul matrigel (BD Bioscience), and injected into the subcutaneous tissues of mouse right lower limbs. Animals were grouped randomly (eight mouse per group) and medication was initiated when the average xenograft size was over 100 mm3: Oxaliplatin was given alone weekly through intraperitoneal injection (5 mg/kg), or in combination with liproxstatin-1 through intraperitoneal injection for twice a week (125 mg/kg) and RSL3 through intra-tumor injection (100 mg/kg, in order to achieve better local concentration and reduce the probable systemic toxicity of RSL3) weekly.

    Click to Show/Hide
Response regulation KIF20A was highly expressed in the oxaliplatin-resistant cell lines and was strongly correlated with survival among colorectal cancer patients. Silencing KIF20A enhanced cellular sensitivity to oxaliplatin both in vivo and in vitro, and silencing KIF20A also suppressed NUAK1 activation. Moreover, silencing NUAK1 up-regulated the expression of PP1, down-regulated the phosphorylation of downstream GSK3Ser9, suppressed the nuclear import of Nrf2, inhibited the expression of a ferroptosis key negative regulatory protein (GPX4), and blocked cellular resistance.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [22]
Target for Ferroptosis Marker/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator LINC00239 (IncRNA) Suppressor
Pathway Response Pathways in cancer hsa05200
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model RKO cells Colon carcinoma Homo sapiens CVCL_0504
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
FHC cells Normal Homo sapiens CVCL_3688
In Vivo Model
To clarify the role of LINC00239 in vivo, we used 4-week-old male BALB/c nude mice provided by the Experimental Animal Center of the Air Force Military Medical University. HCT116 or SW620 cells (1 x 107 cells) were injected subcutaneously into the right flanks of these mice to establish a CRC xenograft model. One week after the injection of cells, the volume of xenografts was continuously monitored (once a week). Four weeks later, the xenografts were removed, and the weights were measured.

    Click to Show/Hide
Response regulation LINC00239 plays a novel and indispensable role in ferroptosis by nucleotides 1-315 of LINC00239 to interact with the Kelch domain (Nrf2-binding site) of Keap1, inhibiting Nrf2 ubiquitination and increasing Nrf2 protein stability. And LINC00239 expression has a positive correlation with Nrf2 and GPX4 expression in CRC tissues. LINC00239 inhibition in combination with ferroptosis induction might be a promising therapeutic strategy for colorectal cancer patients.
Long-chain-fatty-acid--CoA ligase 4 (ACSL4)
In total 10 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [4]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Apatinib Investigative
Responsed Regulator Elongation of very long chain fatty acids protein 6 (ELOVL6) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
HIEC-6 cells Normal Homo sapiens CVCL_6C21
Response regulation ACSL4, a vital regulator of ferroptosis, could interact with ELOVL6 directly. Apatinib promoted ferroptosis in colorectal cancer (CRC) cells by targeting ELOVL6/ACSL4, providing a new mechanism support for apatinib application in the clinical treatment of CRC.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator hsa-miR-19b-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator hsa-miR-130a-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator hsa-miR-150-5p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator hsa-miR-144-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator hsa-miR-16-5p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator mmu-miR-7a-5p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator hsa-miR-17-5p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [5]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Bromelain Investigative
Responsed Regulator hsa-miR-130a-3p (miRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model NCI-H508 cells Cecum adenocarcinoma Homo sapiens CVCL_1564
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
G13D (Human colorectal cancer cells)
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
G12D (Human colorectal cancer cells)
CCD-18Co cells Normal Homo sapiens CVCL_2379
In Vivo Model
Animals (n = 7) were given 2.5% DSS in drinking water for 5 days and then no treatment for 14 days as one cycle; this process was repeated for three cycles. In the last cycle, 2% DSS water treated to each group and no treatment for 14 days. During the three DSS cycle, 3 mg/kg bromelain were injected daily intraperitoneally and colon and spleen tissues were harvested after three DSS cycle in 57 days to study polyp burden and to perform histological staining.

    Click to Show/Hide
Response regulation Elevated miR-19b-3p, -130a-3p, -150-5p, -144-3p, -16-5p, -7a-5p, and -17-5p in bromelain-treated CaCO2cells compared to in DLD-1 cells potentially targeted ACSL-4 and resulted in suppression of ACSL-4. Overall, bromelain inhibits proliferation of Kras mutant Colorectal Cancer (CRC) effectively via ACSL-4.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [20]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator WD repeat domain phosphoinositide-interacting protein 2 (WIPI2) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
HT29 cells Colon cancer Mus musculus CVCL_A8EZ
Response regulation The expression level of ACSL4 decreased and that of GPX4 increased when WIPI2 was knocked down, suggesting that WIPI2 can potentially positively regulate colorectal cancer ferroptosis.
Heme oxygenase 1 (HMOX1)
In total 5 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [2]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Cetuximab Approved
Responsed Regulator Mitogen-activated protein kinase 14 (MAPK14) Driver
Pathway Response Ferroptosis hsa04216
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
The DLD-1 cell suspension (4 x 106 cells/200 ul) was injected subcutaneously into the right dorsal flank of 5-week-old male BALB/c nude mice (Charles River, China). The mice were randomly divided into four groups (5 mice/group): 1) the control group, 2) the RSL3 group, 3) the cetuximab group, and 4) the RSL3 + cetuximab group. Both RSL3 (5 mg/kg) and cetuximab (13 mg/kg) were administered by intraperitoneal injection in a volume of 100 ul once per day. The tumour volume was calculated as 0.5 x length x width2. After 17 days of treatment, the mice were sacrificed, and the tumours were removed. Then, tumour tissue obtained from the different treated groups was subjected to western blotting and immunohistochemical experiments.

    Click to Show/Hide
Response regulation Our work reveals that cetuximab enhances the cytotoxic effect of RSL3 on KRAS mutant Colorectal cancer (CRC) cells and that cetuximab enhances RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 axis through the activation of p38 MAPK.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [29]
Target for Ferroptosis Driver/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Andrographis Approved
Pathway Response Wnt signaling pathway hsa04310
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
Seven-week-old male athymic nude mice (Envigo, Houston, TX) were housed under controlled conditions of light and fed ad libitum. Approximately 5 x 106 parental and 5FUR HCT116 cells were suspended in the matrigel matrix (BD Biosciences, Franklin Lakes, NJ) and subcutaneously injected into mice using a 27-gauge needle (n = 10 per group). Mice were randomly assigned to different treatment groups and 5FU (30 mg/kg body weight) or andrographis (125 mg/kg body weight) or their combination were given intraperitoneally on alternative days for up to 15 days.

    Click to Show/Hide
Response regulation Combined treatment with andrographis was significantly more effective than 5FU and andrographis alone and that these effects were in part orchestrated through dysregulated expression of key genes (including HMOX1, GCLC, GCLM and TCF7L2) within the ferroptosis and Wnt-signaling pathways. Andrographis might offer a safe and inexpensive adjunctive therapeutic option in the management of colorectal cancer patients.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Propofol Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HT29 cells Colon cancer Mus musculus CVCL_A8EZ
CT26 cells Colon adenocarcinoma Mus musculus CVCL_7254
In Vivo Model
CT26 (1 x 105 cells/100 uL) were injected into thetail veinof male BALB/c mice. Then the mice were randomly divided into saline, vehicle, propofol, and sevoflurane groups (n = 5 per group). Saline, fat emulsion (as vehicle control of propofol), and propofol (200 mg/kg) were intraperitoneally injected, while sevoflurane (1.8-2.0%) was administered by inhalation for 2 h. In another set of experiments, coloncancer cells (CT26 and HT29) were pretreated with two doses of propofol (5 ug/mL, 10 ug/mL) or fat emulsion (as vehicle control of propofol) in a cell culture medium for 2 h. After washing with phosphate-buffered saline (PBS), the cells were harvested,counted on a hemacytometer and prepared. Cells (CT26: 1 x 105 cells/100 uL, HT29: 1 x 106 cells/100 uL) were finnally injected into mice through the tail vein.Lung metastasiswas detected via hematoxylin and eosin staining (HE) or ex vivo bioluminescence imaging.

    Click to Show/Hide
Response regulation Further studies showed that propofol treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target genes, including HO-1, NQO1, and SLC7A11. Collectively, we demonstrated the risk of a specific type of anesthetic, propofol, in promoting colorectal cancer cell metastasis through Nrf2-mediated ferroptosis inhibition.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [25]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug tagitinin C Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Response regulation Tagitinin C induces ferroptosis in colorectal cancer cells and has synergistic effect together with erastin. Mechanistically, tagitinin C induces ferroptosis through ER stress-mediated activation of PERK-Nrf2-HO-1 signaling pathway.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [28]
Target for Ferroptosis Driver/Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Talaroconvolutin A Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
In Vivo Model
5 x 106 HCT116 cells were inoculated subcutaneously in the underarm of Balb/c nude female mice (5-week old). The inoculated mice were randomly divided into two groups (6 mice each group). When the tumor reached 300 mm3, the drug group was given TalaA intraperitoneally at a dose of 6.0 mg/kg, and the control group was given the same dose of cosolvent-corn oil. The drug (or cosolvent) was injected every 2 days. Body weight and tumor volume were measured every 2 days.

    Click to Show/Hide
Response regulation Talaroconvolutin A (TalaA) downregulated the expression of the channel protein solute carrier family 7 member 11 (SLC7A11) but upregulated arachidonate lipoxygenase 3 (ALOXE3), promoting ferroptosis. TalaA causes upregulation of HMOX1 which lead to the degradation of heme and the release of free iron, accumulating in mitochondria and giving rise to lipid peroxidation. TalaA could be a new potential powerful drug candidate for colorectal cancer therapy.
Cystine/glutamate transporter (SLC7A11)
In total 6 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [6]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Metformin Investigative
Responsed Regulator Protein FAM98A (FAM98A) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model RKO cells Colon carcinoma Homo sapiens CVCL_0504
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
HCT 15 cells Colon adenocarcinoma Homo sapiens CVCL_0292
DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
FHC cells Normal Homo sapiens CVCL_3688
In Vivo Model
A total 1 x 106 SW620-vector or SW620-FAM98A cells were suspended in 100 ul PBS and injected s.c. into the back of 4- to 6-week-old male BALB/cnude mice (Laboratory Animal Unit, Southern Medical University, China). The sizes of the resulting tumors were measured weekly. Tumor volumes were calculated as follows: total tumor volume (mm3) = (Length x Width2)/2, where Length is the longest length. When the tumor sizes reached about 200 mm3, nude mice in the four groups were given PBS or 5-FU treatment (30 mg/kg, intraperitoneal injection, twice a week), respectively. Nude mice were maintained in a barrier facility in racks filtered with a high-efficiency particulate air filter.

    Click to Show/Hide
Response regulation The expressions of FAM98A and SLC7A11 were also downregulated after metformin treatment. And FAM98A is predominantly expressed in the colorectal cancer tissues and high FAM98A expression is usually accompanied by the high expression of SLC7A11, which usually means ferroptosis resistance.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [7]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Niraparib Investigative
Responsed Regulator Cyclic GMP-AMP synthase (CGAS) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cytosolic DNA-sensing pathway hsa04623
Cell Process Cell ferroptosis
In Vitro Model HT29 cells Colon cancer Mus musculus CVCL_A8EZ
CT26 cells Colon adenocarcinoma Mus musculus CVCL_7254
MC-38 cells Colon adenocarcinoma Homo sapiens CVCL_B288
In Vivo Model
Six-week-old male BALB/c athymic nude mice were purchased from the Experimental Animal Center of Peking (Beijing, China). Stable cells (5 x 106) were seeded into the right flanks of the mice. After the xenografts had grown to 200 mm3, saline as a vehicle or sorafenib (30 mg/kg) was administered by gavage every day, and the mice were euthanized by the cervical dislocation method five weeks later. Before sacrifice, the tumor sizes and body weights were measured twice per week. The tumor volume (V) was calculated as follows: (L x W2)/2 (length, L, and width, W). The xenografts were excised and further assessed.

    Click to Show/Hide
Response regulation Niraparib, a widely used PARPi, augmented cGAS-mediated ferroptosis and immune activation. In colorectal cancer models, cGAS signaling exerts tumor control via ATF3SLC7A11GPX4-mediated ferroptosis and IFNCD8 T cell-mediated antitumor immune response.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [8]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Sodium butyrate Investigative
Responsed Regulator CD44 antigen (CD44) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model FHC cells Normal Homo sapiens CVCL_3688
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
In Vivo Model
Six-week-old male C57BL/6J mice were purchased from the Medical Laboratory Animal Center of Guangdong Province (Foshan, China). Forty-five C57BL/6L mice were randomized into 4 groups after 1 week of adaptive feeding: (1) Control group (n = 9); (2) AOM/DSS group (n = 12); (3) AOM/DSS + NaB (orally) group (n = 12); 4) AOM/DSS + NaB (intraperitoneal injection) group (n = 12). The Control group received an intraperitoneal injection of saline solution beginning on day 1, and received sterile drinking water throughout the study. Other three groups received an intraperitoneal injection of 10 mg/kg AOM (Sigma Aldrich) beginning on day 1, and received drinking water containing 2.5% DSS at the second and eighth weeks (2% DSS in the fifth week). Besides, 0.1 M NaB (Sigma Aldrich) was given in drinking water during the whole experiment process in AOM/DSS + NaB (p.o.) group, while AOM/DSS + NaB (i.p.) group was injected intraperitoneally (IP) with 1 g/kg NaB per day.

    Click to Show/Hide
Response regulation Sodium butyrate (NaB) induces ferroptosis in colorectal cancer cells through the CD44/SLC7A11 signaling pathway and has synergistic effects with Erastin.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [30]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug 2-Imino-6-methoxy-2H-chromene-3-carbothioamide Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
mTOR signaling pathway hsa04150
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model DLD-1 cells Colon adenocarcinoma Homo sapiens CVCL_0248
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
In Vivo Model
Five-week-old female BALB/c nude mice were purchased from Beijing Weitong Lihua Experimental Animal Technical Co., Ltd. (Beijing, China). The mice were randomly assigned to the treatment and control groups until the tumor size reached approximately 100 mm3. The mice in the treatment group were injected with 0.174 mg/mL IMCA (100 uL), and those in the control group were injected with an equal volume of normal saline. The nude mice were euthanized, and samples were obtained from their tumor, heart, hepar, kidney, and blood after 33 days of IMCA treatment.

    Click to Show/Hide
Response regulation 2-imino-6-methoxy-2H-chromene-3-carbothioamide (IMCA) significantly induced the ferroptosis of colorectal cancer cells. Mechanistically, IMCA downregulated the expression of SLC7A11 and decreased the contents of cysteine and glutathione, which resulted in reactive oxygen species accumulation and ferroptosis.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Propofol Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HT29 cells Colon cancer Mus musculus CVCL_A8EZ
CT26 cells Colon adenocarcinoma Mus musculus CVCL_7254
In Vivo Model
CT26 (1 x 105 cells/100 uL) were injected into thetail veinof male BALB/c mice. Then the mice were randomly divided into saline, vehicle, propofol, and sevoflurane groups (n = 5 per group). Saline, fat emulsion (as vehicle control of propofol), and propofol (200 mg/kg) were intraperitoneally injected, while sevoflurane (1.8-2.0%) was administered by inhalation for 2 h. In another set of experiments, coloncancer cells (CT26 and HT29) were pretreated with two doses of propofol (5 ug/mL, 10 ug/mL) or fat emulsion (as vehicle control of propofol) in a cell culture medium for 2 h. After washing with phosphate-buffered saline (PBS), the cells were harvested,counted on a hemacytometer and prepared. Cells (CT26: 1 x 105 cells/100 uL, HT29: 1 x 106 cells/100 uL) were finnally injected into mice through the tail vein.Lung metastasiswas detected via hematoxylin and eosin staining (HE) or ex vivo bioluminescence imaging.

    Click to Show/Hide
Response regulation Further studies showed that propofol treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target genes, including HO-1, NQO1, and SLC7A11. Collectively, we demonstrated the risk of a specific type of anesthetic, propofol, in promoting colorectal cancer cell metastasis through Nrf2-mediated ferroptosis inhibition.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [28]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Talaroconvolutin A Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
In Vivo Model
5 x 106 HCT116 cells were inoculated subcutaneously in the underarm of Balb/c nude female mice (5-week old). The inoculated mice were randomly divided into two groups (6 mice each group). When the tumor reached 300 mm3, the drug group was given TalaA intraperitoneally at a dose of 6.0 mg/kg, and the control group was given the same dose of cosolvent-corn oil. The drug (or cosolvent) was injected every 2 days. Body weight and tumor volume were measured every 2 days.

    Click to Show/Hide
Response regulation Talaroconvolutin A (TalaA) downregulated the expression of the channel protein solute carrier family 7 member 11 (SLC7A11) but upregulated arachidonate lipoxygenase 3 (ALOXE3), promoting ferroptosis. TalaA causes upregulation of HMOX1 which lead to the degradation of heme and the release of free iron, accumulating in mitochondria and giving rise to lipid peroxidation. TalaA could be a new potential powerful drug candidate for colorectal cancer therapy.
Unspecific Target
In total 15 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Auriculasin Investigative
Responsed Regulator Kelch-like ECH-associated protein 1 (KEAP1) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell apoptosis
Cell invasion
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
Response regulation Auriculasin promoted the expression of Keap1 and AIFM1, but significantly reduced the phosphorylation level of AIFM1. AC can promote colorectal cancer cell apoptosis, ferroptosis and oxeiptosis by inducing ROS generation, thereby inhibiting cell viability, invasion and colony formation, indicating that AC has a significant tumor suppressor effect.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [9]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Auriculasin Investigative
Responsed Regulator Apoptosis-inducing factor 1, mitochondrial (AIFM1) Driver
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell apoptosis
Cell invasion
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
Response regulation Auriculasin promoted the expression of Keap1 and AIFM1, but significantly reduced the phosphorylation level of AIFM1. AC can promote colorectal cancer cell apoptosis, ferroptosis and oxeiptosis by inducing ROS generation, thereby inhibiting cell viability, invasion and colony formation, indicating that AC has a significant tumor suppressor effect.
Experiment 3 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug BEBT-908 Investigative
Responsed Regulator Histone deacetylase 1 (HDAC1) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
PI3K-Akt signaling pathway hsa04151
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MC-38 cells Colon adenocarcinoma Homo sapiens CVCL_B288
In Vivo Model
About 2 x 105 MC38 tumor cells were inoculated subcutaneously into C57BL/6 mice. A total of 100 mg/kg BEBT-908 was intravenously injected every other day for four times initiating from day 4 after tumor cell inoculation. Tumors were harvested on day 12 after inoculation, weighted, mechanically minced and incubated with 50 ug/mL DNase I (Sigma) and 2 mg/mL collagenase P (Sigma) for 20 minutes at 37.

    Click to Show/Hide
Response regulation Treatment with BEBT-908 promoted ferroptotic cell death of cancer cells by hyperacetylating p53 and facilitating the expression of ferroptotic signaling. The dual PI3K/HDAC inhibitor BEBT-908 elicits potent antitumor responses, effectively inducing immunogenic ferroptosis of colorectal cancer cells and potentiating cancer immunotherapy.
Experiment 4 Reporting the Ferroptosis-centered Disease Response by This Target [10]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug BEBT-908 Investigative
Responsed Regulator Cellular tumor antigen p53 (TP53) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
PI3K-Akt signaling pathway hsa04151
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model MC-38 cells Colon adenocarcinoma Homo sapiens CVCL_B288
In Vivo Model
About 2 x 105 MC38 tumor cells were inoculated subcutaneously into C57BL/6 mice. A total of 100 mg/kg BEBT-908 was intravenously injected every other day for four times initiating from day 4 after tumor cell inoculation. Tumors were harvested on day 12 after inoculation, weighted, mechanically minced and incubated with 50 ug/mL DNase I (Sigma) and 2 mg/mL collagenase P (Sigma) for 20 minutes at 37.

    Click to Show/Hide
Response regulation Treatment with BEBT-908 promoted ferroptotic cell death of cancer cells by hyperacetylating p53 and facilitating the expression of ferroptotic signaling. The dual PI3K/HDAC inhibitor BEBT-908 elicits potent antitumor responses, effectively inducing immunogenic ferroptosis of colorectal cancer cells and potentiating cancer immunotherapy.
Experiment 5 Reporting the Ferroptosis-centered Disease Response by This Target [31]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Ascorbic Acid Approved
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model DiFi cells Colorectal carcinoma Homo sapiens CVCL_6895
CCK-81 cells Colon adenocarcinoma Homo sapiens CVCL_2873
C75 cells Colon adenocarcinoma Homo sapiens CVCL_5248
In Vivo Model
The PDX models used in this study were derived from patients (CRC0078 and CRC0121) carrying a quadruple wild-type (KRAS,NRAS,BRAF, andPIK3CA) colorectal tumor. Established tumors (average volume 300 or 500 mm3, as indicated) were treated with the following regimens, either single-agent or in combination: VitC (Sigma, 4 g/kg, intraperitoneal, daily-5 days per week), cetuximab (Merck, 10 mg/kg, intraperitoneal, twice weekly).

    Click to Show/Hide
Response regulation Cetuximab blunts carbohydrate metabolism by blocking glucose uptake and glycolysis, beyond promoting slow but progressive ROS production. In parallel, Vitamin C disrupts iron homeostasis and further increases ROS levels ultimately leading to ferroptosis. Considering that high-dose VitC is known to be safe in cancer patients, this findings might have clinical impact on colorectal cancer patients treated with anti-EGFR therapies.
Experiment 6 Reporting the Ferroptosis-centered Disease Response by This Target [31]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Cetuximab Approved
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model DiFi cells Colorectal carcinoma Homo sapiens CVCL_6895
CCK-81 cells Colon adenocarcinoma Homo sapiens CVCL_2873
C75 cells Colon adenocarcinoma Homo sapiens CVCL_5248
In Vivo Model
The PDX models used in this study were derived from patients (CRC0078 and CRC0121) carrying a quadruple wild-type (KRAS,NRAS,BRAF, andPIK3CA) colorectal tumor. Established tumors (average volume 300 or 500 mm3, as indicated) were treated with the following regimens, either single-agent or in combination: VitC (Sigma, 4 g/kg, intraperitoneal, daily-5 days per week), cetuximab (Merck, 10 mg/kg, intraperitoneal, twice weekly).

    Click to Show/Hide
Response regulation Cetuximab blunts carbohydrate metabolism by blocking glucose uptake and glycolysis, beyond promoting slow but progressive ROS production. In parallel, Vitamin C disrupts iron homeostasis and further increases ROS levels ultimately leading to ferroptosis. Considering that high-dose VitC is known to be safe in cancer patients, this findings might have clinical impact on colorectal cancer patients treated with anti-EGFR therapies.
Experiment 7 Reporting the Ferroptosis-centered Disease Response by This Target [32]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Cetuximab Approved
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell adhesion molecules hsa04514
Cell Process Cell ferroptosis
Cell proliferation
Cell migration
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
In Vivo Model
5-week-old female BALB/c nude were purchased from Shanghai Slac Laboratory Animal Co., Ltd. HCT116-luc cells were digested and washed by cold PBS for three times, and the final concentration was 2.5 x 106/ml in cold PBS. A volume of 100 ul cell suspension was injected subcutaneously into right dorsal flank of mice. When the tumor tissue in the subcutaneous was macroscopic, the tumor tissues were dissected and embedded into the mesentery of mice.

    Click to Show/Hide
Response regulation b-elemene in combination with cetuximab was shown to induce iron-dependent reactive oxygen species (ROS) accumulation, glutathione (GSH) depletion, lipid peroxidation, upregulation of HO-1 and transferrin, and downregulation of negative regulatory proteins for ferroptosis (GPX4, SLC7A11, FTH1, glutaminase, and SLC40A1) in KRAS mutant colorectal cancer cells.
Experiment 8 Reporting the Ferroptosis-centered Disease Response by This Target [32]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Beta-Elemene Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell adhesion molecules hsa04514
Cell Process Cell ferroptosis
Cell proliferation
Cell migration
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
Caco-2 cells Colon adenocarcinoma Homo sapiens CVCL_0025
In Vivo Model
5-week-old female BALB/c nude were purchased from Shanghai Slac Laboratory Animal Co., Ltd. HCT116-luc cells were digested and washed by cold PBS for three times, and the final concentration was 2.5 x 106/ml in cold PBS. A volume of 100 ul cell suspension was injected subcutaneously into right dorsal flank of mice. When the tumor tissue in the subcutaneous was macroscopic, the tumor tissues were dissected and embedded into the mesentery of mice.

    Click to Show/Hide
Response regulation b-elemene in combination with cetuximab was shown to induce iron-dependent reactive oxygen species (ROS) accumulation, glutathione (GSH) depletion, lipid peroxidation, upregulation of HO-1 and transferrin, and downregulation of negative regulatory proteins for ferroptosis (GPX4, SLC7A11, FTH1, glutaminase, and SLC40A1) in KRAS mutant colorectal cancer cells.
Experiment 9 Reporting the Ferroptosis-centered Disease Response by This Target [33]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Dichloroacetate Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell adhesion molecules hsa04514
Cell Process Cell ferroptosis
Cell migration
Cell stemness
In Vitro Model HT-29 cells Colon adenocarcinoma Homo sapiens CVCL_0320
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Response regulation Dichloroacetate (DCA) could sequester iron in lysosome and thus trigger ferroptosis, which is necessary for DCA-mediated attenuation on colorectal cancer cell stemness. And DCA can attenuate the stemness of colorectal CSCs without affecting the viability of colorectal cancer cells.
Experiment 10 Reporting the Ferroptosis-centered Disease Response by This Target [34]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Punicic Acid Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model FaDu cells Hypopharyngeal squamous cell carcinoma Homo sapiens CVCL_1218
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
Response regulation Punicic acid is highly cytotoxic to HCT-116 colorectal carcinoma and FaDu hypopharyngeal carcinoma cells grown either in monolayers or as three-dimensional spheroids. It induces significant lipid peroxidation and its effects are prevented by the addition of ferroptosis inhibitors.
Experiment 11 Reporting the Ferroptosis-centered Disease Response by This Target [35]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug SN38 Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
In Vitro Model Colon 26-L5 cells Colon adenocarcinoma Mus musculus CVCL_3924
Response regulation The anticancer effect of the combined treatment of SN38 and EP is related to changes in the redox-homeostasis of cancer cells, leading to cell death via apoptosis and/or ferroptosis. Thus, electroporation has a potential to increase the sensitivity of colorectal cancer cells to conventional anticancer therapy with SN38.
Experiment 12 Reporting the Ferroptosis-centered Disease Response by This Target [36]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Circ_0007142 (circRNA) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
FHC cells Normal Homo sapiens CVCL_3688
In Vivo Model
3 x 106 HCT116 cells with stable transfection of lentiviral vectors sh-circ_0007142 or sh-NC were harvested and resuspended in 0.2 mL PBS. Six-week-old male BALB/c nude mice (n = 12) from Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China) were injected with the prepared cell suspension (6 mice/group). During the 4-week observation period, tumour volume (length x width2/2) was detected every week.

    Click to Show/Hide
Response regulation Circ_0007142 regulated cancer progression and ferroptosis in colorectal cancer (CRC) cells partly by acting on the miR-874-3p/GDPD5 axis. This is a specific regulatory network for ferroptosis in CRC, and CRC treatment might be improved with circ_0007142 as a therapeutic target aiming at ferroptosis.
Experiment 13 Reporting the Ferroptosis-centered Disease Response by This Target [36]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator hsa-miR-874-3p (miRNA) Driver
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Apoptosis hsa04210
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
FHC cells Normal Homo sapiens CVCL_3688
In Vivo Model
3 x 106 HCT116 cells with stable transfection of lentiviral vectors sh-circ_0007142 or sh-NC were harvested and resuspended in 0.2 mL PBS. Six-week-old male BALB/c nude mice (n = 12) from Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China) were injected with the prepared cell suspension (6 mice/group). During the 4-week observation period, tumour volume (length x width2/2) was detected every week.

    Click to Show/Hide
Response regulation Circ_0007142 regulated cancer progression and ferroptosis in colorectal cancer (CRC) cells partly by acting on the miR-874-3p/GDPD5 axis. This is a specific regulatory network for ferroptosis in CRC, and CRC treatment might be improved with circ_0007142 as a therapeutic target aiming at ferroptosis.
Experiment 14 Reporting the Ferroptosis-centered Disease Response by This Target [14]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Tumor necrosis factor alpha-induced protein 8 (TNFAIP8) Suppressor
Pathway Response Ferroptosis hsa04216
MAPK signaling pathway hsa04010
Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
HEK-293T cells Normal Homo sapiens CVCL_0063
In Vivo Model
Six 4-week-old male BALB/c nude mice were ordered from the Shanghai Laboratory Animal Center (Shanghai SLAC Laboratory Animal Co., Ltd., China). A total of 5 x 106 TIPE+/+ SW480 cells were suspended in 100 uL of PBS and subcutaneously injected into the right axilla flank of each nude mouse, and the same amount of vector SW480 cells was into the left. At 2 weeks after inoculation, the xenograft tumor size was measured using Vernier calipers every 2 days.

    Click to Show/Hide
Response regulation MiR-539 can bind to and regulate the expression of TIPE, and miR-539 activates SAPK/JNK to downregulate the expression of glutathione peroxidase 4 (GPX4) and promote ferroptosis. In addition, SAPK/JNK is the upstream molecule of p53. MiR-539 is a new therapeutic target for colorectal cancer (CRC) patients.
Experiment 15 Reporting the Ferroptosis-centered Disease Response by This Target [37]
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator Metallothionein-1G (MT1G) Suppressor
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT-8 cells Ileocecal adenocarcinoma Homo sapiens CVCL_2478
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
NCM460 cells Normal Homo sapiens CVCL_0460
Response regulation The downregulated ferroptosis-associated gene MT1G and revealed that a low MT1G level displayed a worse prognosis in colorectal cancer patients. In addition, the MT1G expression was closely related to the immune microenvironment and involved in the progression of tumors.
Serotransferrin (TF)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [11]
Target for Ferroptosis Marker/Suppressor/Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator hsa-miR-545-3p (miRNA) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
In Vitro Model NCM460 cells Normal Homo sapiens CVCL_0460
HT-29 cells Colon adenocarcinoma Homo sapiens CVCL_0320
HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
LoVo cells Colon adenocarcinoma Homo sapiens CVCL_0399
In Vivo Model
5-week-old immunodeficient nude mice (male, weight, 16-20 g, n = 40 mice for each group) were purchased from Cyagen bio. Co. (Beijing, China). Before experiments, the mice were adapted to the breeding environment for two weeks. All mice were maintained at a 12 h/12 h light/dark cycle with free access to water and food. A total of 5 x 106 HT-29 or HCT-116 cells were suspended in 100 uL PBS and injected subcutaneously into the right posterior flanks of nude mice. After three weeks, the mice were killed and the tumor xenografts were dissected, weighed and fixed in 10% buffered formaldehyde for further IHC analysis.

    Click to Show/Hide
Response regulation MiR-545 inhibits ferroptosis in colorectal cancer cells by inhibiting TF. TF overexpression blocked miR-545-induced changes in ROS, MDA, and Fe2+ levels in HT-29 and HCT-116 cells, thereby inducing CRC cell death.
NAD(P)H dehydrogenase [quinone] 1 (NQO1)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [24]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Propofol Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
In Vitro Model HT29 cells Colon cancer Mus musculus CVCL_A8EZ
CT26 cells Colon adenocarcinoma Mus musculus CVCL_7254
In Vivo Model
CT26 (1 x 105 cells/100 uL) were injected into thetail veinof male BALB/c mice. Then the mice were randomly divided into saline, vehicle, propofol, and sevoflurane groups (n = 5 per group). Saline, fat emulsion (as vehicle control of propofol), and propofol (200 mg/kg) were intraperitoneally injected, while sevoflurane (1.8-2.0%) was administered by inhalation for 2 h. In another set of experiments, coloncancer cells (CT26 and HT29) were pretreated with two doses of propofol (5 ug/mL, 10 ug/mL) or fat emulsion (as vehicle control of propofol) in a cell culture medium for 2 h. After washing with phosphate-buffered saline (PBS), the cells were harvested,counted on a hemacytometer and prepared. Cells (CT26: 1 x 105 cells/100 uL, HT29: 1 x 106 cells/100 uL) were finnally injected into mice through the tail vein.Lung metastasiswas detected via hematoxylin and eosin staining (HE) or ex vivo bioluminescence imaging.

    Click to Show/Hide
Response regulation Further studies showed that propofol treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target genes, including HO-1, NQO1, and SLC7A11. Collectively, we demonstrated the risk of a specific type of anesthetic, propofol, in promoting colorectal cancer cell metastasis through Nrf2-mediated ferroptosis inhibition.
Iron-responsive element-binding protein 2 (IREB2)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [27]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Regulator hsa-mir-19a (Precursor RNA) Suppressor
Pathway Response Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HT29 cells Colon cancer Mus musculus CVCL_A8EZ
Response regulation IREB2 was negatively regulated by miR-19a in Colorectal cancer (CRC) cells. In addition, ferroptosis was suppressed by miR-19a through inhibiting IREB2.
Hydroperoxide isomerase ALOXE3 (ALOXE3)
In total 1 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [28]
Target for Ferroptosis Driver
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Talaroconvolutin A Investigative
Pathway Response Fatty acid metabolism hsa01212
Ferroptosis hsa04216
Cell Process Cell ferroptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
SW620 cells Colon adenocarcinoma Homo sapiens CVCL_0547
In Vivo Model
5 x 106 HCT116 cells were inoculated subcutaneously in the underarm of Balb/c nude female mice (5-week old). The inoculated mice were randomly divided into two groups (6 mice each group). When the tumor reached 300 mm3, the drug group was given TalaA intraperitoneally at a dose of 6.0 mg/kg, and the control group was given the same dose of cosolvent-corn oil. The drug (or cosolvent) was injected every 2 days. Body weight and tumor volume were measured every 2 days.

    Click to Show/Hide
Response regulation Talaroconvolutin A (TalaA) downregulated the expression of the channel protein solute carrier family 7 member 11 (SLC7A11) but upregulated arachidonate lipoxygenase 3 (ALOXE3), promoting ferroptosis. TalaA causes upregulation of HMOX1 which lead to the degradation of heme and the release of free iron, accumulating in mitochondria and giving rise to lipid peroxidation. TalaA could be a new potential powerful drug candidate for colorectal cancer therapy.
Ferroptosis suppressor protein 1 (AIFM2)
In total 2 item(s) under this target
Experiment 1 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Andrographis Approved
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
Briefly, surgically resected tumors were maintained in DMEM-F12 (Gibco) supplemented with 1% HEPES (Sigma-Aldrich), 1% L-glutamine (Gibco), 10% FBS (Gibco), 2% penicillin/streptomycin (Sigma-Aldrich), and 10 uM Y-27632 (R&D Systems). Tumors were digested with collagenase solution (5 mL of the above medium with 75 uL collagenase, 124 ug/mL dispase type II, and 0.2% Primocen) for 30 min and then filtered through a 70 um filter (Corning). An organoid pellet was obtained by centrifugation (200x g for 10 min). Organoids were suspended in Matrigel (Corning, Tehama County, CA) with IntestiCult Organoid Growth Medium (#06010, STEMCELL Technologies) and seeded in 12-well plates. Approximately 750 uL of IntestiCult Organoid Growth Medium was added to each well. Organoids were divided into five groups of control, curcumin (3.0 ug/mL), andrographis (30.0 ug/mL), their combination (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL), and their combination plus ferrostatin-1 (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL; ferrostatin-1; 20 uM). Following forty-eight hours of treatment, the numbers of organoids (<100 um) and their mean sizes were examined using Image J software.

    Click to Show/Hide
Response regulation In conclusion, our study revealed that combined treatment with curcumin and andrographis exhibited anti-tumorigenic effects in colorectal cancer cells through activation of ferroptosis and by dual suppression of GPX-4 and FSP-1, which have significant potential implications for the adjunctive treatment of CRC patients. This combination treatment resulted in cancer cell death via both forms of cell death: apoptosis and ferroptosis.
Experiment 2 Reporting the Ferroptosis-centered Disease Response by This Target [12]
Target for Ferroptosis Suppressor
Responsed Disease Colorectal cancer [ICD-11: 2B91]
Responsed Drug Curcumin Investigative
Pathway Response Fatty acid metabolism hsa01212
Cell Process Cell ferroptosis
Cell apoptosis
Cell proliferation
In Vitro Model HCT 116 cells Colon carcinoma Homo sapiens CVCL_0291
SW480 cells Colon adenocarcinoma Homo sapiens CVCL_0546
In Vivo Model
Briefly, surgically resected tumors were maintained in DMEM-F12 (Gibco) supplemented with 1% HEPES (Sigma-Aldrich), 1% L-glutamine (Gibco), 10% FBS (Gibco), 2% penicillin/streptomycin (Sigma-Aldrich), and 10 uM Y-27632 (R&D Systems). Tumors were digested with collagenase solution (5 mL of the above medium with 75 uL collagenase, 124 ug/mL dispase type II, and 0.2% Primocen) for 30 min and then filtered through a 70 um filter (Corning). An organoid pellet was obtained by centrifugation (200x g for 10 min). Organoids were suspended in Matrigel (Corning, Tehama County, CA) with IntestiCult Organoid Growth Medium (#06010, STEMCELL Technologies) and seeded in 12-well plates. Approximately 750 uL of IntestiCult Organoid Growth Medium was added to each well. Organoids were divided into five groups of control, curcumin (3.0 ug/mL), andrographis (30.0 ug/mL), their combination (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL), and their combination plus ferrostatin-1 (curcumin; 3.0 ug/mL, andrographis; 30.0 ug/mL; ferrostatin-1; 20 uM). Following forty-eight hours of treatment, the numbers of organoids (<100 um) and their mean sizes were examined using Image J software.

    Click to Show/Hide
Response regulation In conclusion, our study revealed that combined treatment with curcumin and andrographis exhibited anti-tumorigenic effects in colorectal cancer cells through activation of ferroptosis and by dual suppression of GPX-4 and FSP-1, which have significant potential implications for the adjunctive treatment of CRC patients. This combination treatment resulted in cancer cell death via both forms of cell death: apoptosis and ferroptosis.
References
Ref 1 Curcumin Represses Colorectal Cancer Cell Proliferation by Triggering Ferroptosis via PI3K/Akt/mTOR Signaling. Nutr Cancer. 2023;75(2):726-733. doi: 10.1080/01635581.2022.2139398. Epub 2022 Nov 8.
Ref 2 Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021 Nov 13;12(11):1079. doi: 10.1038/s41419-021-04367-3.
Ref 3 Ibrutinib facilitates the sensitivity of colorectal cancer cells to ferroptosis through BTK/NRF2 pathway. Cell Death Dis. 2023 Feb 23;14(2):151. doi: 10.1038/s41419-023-05664-9.
Ref 4 Apatinib Promotes Ferroptosis in Colorectal Cancer Cells by Targeting ELOVL6/ACSL4 Signaling. Cancer Manag Res. 2021 Feb 11;13:1333-1342. doi: 10.2147/CMAR.S274631. eCollection 2021.
Ref 5 Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim Cells Syst (Seoul). 2018 Aug 30;22(5):334-340. doi: 10.1080/19768354.2018.1512521. eCollection 2018.
Ref 6 FAM98A promotes resistance to 5-fluorouracil in colorectal cancer by suppressing ferroptosis. Arch Biochem Biophys. 2022 Jun 15;722:109216. doi: 10.1016/j.abb.2022.109216. Epub 2022 Apr 11.
Ref 7 PARPi treatment enhances radiotherapy-induced ferroptosis and antitumor immune responses via the cGAS signaling pathway in colorectal cancer. Cancer Lett. 2022 Dec 1;550:215919. doi: 10.1016/j.canlet.2022.215919. Epub 2022 Sep 16.
Ref 8 Sodium Butyrate Induces CRC Cell Ferroptosis via the CD44/SLC7A11 Pathway and Exhibits a Synergistic Therapeutic Effect with Erastin. Cancers (Basel). 2023 Jan 9;15(2):423. doi: 10.3390/cancers15020423.
Ref 9 Auriculasin enhances ROS generation to regulate colorectal cancer cell apoptosis, ferroptosis, oxeiptosis, invasion and colony formation. Biochem Biophys Res Commun. 2022 Jan 8;587:99-106. doi: 10.1016/j.bbrc.2021.11.101. Epub 2021 Dec 1.
Ref 10 A Dual PI3K/HDAC Inhibitor Induces Immunogenic Ferroptosis to Potentiate Cancer Immune Checkpoint Therapy. Cancer Res. 2021 Dec 15;81(24):6233-6245. doi: 10.1158/0008-5472.CAN-21-1547. Epub 2021 Oct 28.
Ref 11 miR-545 promotes colorectal cancer by inhibiting transferring in the non-normal ferroptosis signaling. Aging (Albany NY). 2021 Dec 26;13(24):26137-26147. doi: 10.18632/aging.203801. Epub 2021 Dec 26.
Ref 12 Curcumin and Andrographis Exhibit Anti-Tumor Effects in Colorectal Cancer via Activation of Ferroptosis and Dual Suppression of Glutathione Peroxidase-4 and Ferroptosis Suppressor Protein-1. Pharmaceuticals (Basel). 2023 Mar 2;16(3):383. doi: 10.3390/ph16030383.
Ref 13 RSL3 Drives Ferroptosis Through GPX4 Inactivation and ROS Production in Colorectal Cancer. Front Pharmacol. 2018 Nov 22;9:1371. doi: 10.3389/fphar.2018.01371. eCollection 2018.
Ref 14 miR-539 activates the SAPK/JNK signaling pathway to promote ferropotosis in colorectal cancer by directly targeting TIPE. Cell Death Discov. 2021 Oct 2;7(1):272. doi: 10.1038/s41420-021-00659-x.
Ref 15 TIMP1 represses sorafenib-triggered ferroptosis in colorectal cancer cells by activating the PI3K/Akt signaling pathway. Immunopharmacol Immunotoxicol. 2023 Dec;45(4):419-425. doi: 10.1080/08923973.2022.2160731. Epub 2023 Mar 12.
Ref 16 HSPA5 repressed ferroptosis to promote colorectal cancer development by maintaining GPX4 stability. Neoplasma. 2022 Sep;69(5):1054-1069. doi: 10.4149/neo_2022_220331N363. Epub 2022 Jun 20.
Ref 17 Development of a prognostic model based on ferroptosis-related genes for colorectal cancer patients and exploration of the biological functions of NOS2 in vivo and in vitro. Front Oncol. 2023 Jun 6;13:1133946. doi: 10.3389/fonc.2023.1133946. eCollection 2023.
Ref 18 ACADSB regulates ferroptosis and affects the migration, invasion, and proliferation of colorectal cancer cells. Cell Biol Int. 2020 Nov;44(11):2334-2343. doi: 10.1002/cbin.11443. Epub 2020 Aug 22.
Ref 19 Inhibition of SRSF9 enhances the sensitivity of colorectal cancer to erastin-induced ferroptosis by reducing glutathione peroxidase 4 expression. Int J Biochem Cell Biol. 2021 May;134:105948. doi: 10.1016/j.biocel.2021.105948. Epub 2021 Feb 17.
Ref 20 WIPI2 enhances the vulnerability of colorectal cancer cells to erastin via bioinformatics analysis and experimental verification. Front Oncol. 2023 May 3;13:1146617. doi: 10.3389/fonc.2023.1146617. eCollection 2023.
Ref 21 MiR-15a-3p regulates ferroptosis via targeting glutathione peroxidase GPX4 in colorectal cancer. Mol Carcinog. 2022 Mar;61(3):301-310. doi: 10.1002/mc.23367. Epub 2021 Nov 2.
Ref 22 Long noncoding RNA LINC00239 inhibits ferroptosis in colorectal cancer by binding to Keap1 to stabilize Nrf2. Cell Death Dis. 2022 Aug 29;13(8):742. doi: 10.1038/s41419-022-05192-y.
Ref 23 Oxaliplatin induces ferroptosis and oxidative stress in HT29 colorectal cancer cells by inhibiting the Nrf2 signaling pathway. Exp Ther Med. 2022 Jun;23(6):394. doi: 10.3892/etm.2022.11321. Epub 2022 Apr 13.
Ref 24 Anesthetic propofol inhibits ferroptosis and aggravates distant cancer metastasis via Nrf2 upregulation. Free Radic Biol Med. 2023 Feb 1;195:298-308. doi: 10.1016/j.freeradbiomed.2022.12.092. Epub 2022 Dec 29.
Ref 25 Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. Int J Biol Sci. 2021 Jun 26;17(11):2703-2717. doi: 10.7150/ijbs.59404. eCollection 2021.
Ref 26 Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin. Aging (Albany NY). 2021 Mar 26;13(10):13515-13534. doi: 10.18632/aging.202774. Epub 2021 Mar 26.
Ref 27 MiR-19a suppresses ferroptosis of colorectal cancer cells by targeting IREB2. Bioengineered. 2022 May;13(5):12021-12029. doi: 10.1080/21655979.2022.2054194.
Ref 28 Discovery of a novel ferroptosis inducer-talaroconvolutin A-killing colorectal cancer cells in vitro and in vivo. Cell Death Dis. 2020 Nov 17;11(11):988. doi: 10.1038/s41419-020-03194-2.
Ref 29 Andrographis-mediated chemosensitization through activation of ferroptosis and suppression of -catenin/Wnt-signaling pathways in colorectal cancer. Carcinogenesis. 2020 Oct 15;41(10):1385-1394. doi: 10.1093/carcin/bgaa090.
Ref 30 IMCA Induces Ferroptosis Mediated by SLC7A11 through the AMPK/mTOR Pathway in Colorectal Cancer. Oxid Med Cell Longev. 2020 Apr 3;2020:1675613. doi: 10.1155/2020/1675613. eCollection 2020.
Ref 31 Vitamin C Restricts the Emergence of Acquired Resistance to EGFR-Targeted Therapies in Colorectal Cancer. Cancers (Basel). 2020 Mar 14;12(3):685. doi: 10.3390/cancers12030685.
Ref 32 Combinative treatment of -elemene and cetuximab is sensitive to KRAS mutant colorectal cancer cells by inducing ferroptosis and inhibiting epithelial-mesenchymal transformation. Theranostics. 2020 Apr 6;10(11):5107-5119. doi: 10.7150/thno.44705. eCollection 2020.
Ref 33 Dichloroacetate attenuates the stemness of colorectal cancer cells via trigerring ferroptosis through sequestering iron in lysosomes. Environ Toxicol. 2021 Apr;36(4):520-529. doi: 10.1002/tox.23057. Epub 2020 Nov 9.
Ref 34 Punicic Acid Triggers Ferroptotic Cell Death in Carcinoma Cells. Nutrients. 2021 Aug 10;13(8):2751. doi: 10.3390/nu13082751.
Ref 35 Redox-related Molecular Mechanism of Sensitizing Colon Cancer Cells to Camptothecin Analog SN38. Anticancer Res. 2020 Sep;40(9):5159-5170. doi: 10.21873/anticanres.14519.
Ref 36 Circ_0007142 downregulates miR-874-3p-mediated GDPD5 on colorectal cancer cells. Eur J Clin Invest. 2021 Jul;51(7):e13541. doi: 10.1111/eci.13541. Epub 2021 Apr 2.
Ref 37 Ferroptosis-Related Gene MT1G as a Novel Biomarker Correlated With Prognosis and Immune Infiltration in Colorectal Cancer. Front Cell Dev Biol. 2022 Apr 20;10:881447. doi: 10.3389/fcell.2022.881447. eCollection 2022.