Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Nature ; 585(7826): 603-608, 2020 09.
Article in English | MEDLINE | ID: mdl-32939090

ABSTRACT

Ferroptosis-an iron-dependent, non-apoptotic cell death process-is involved in various degenerative diseases and represents a targetable susceptibility in certain cancers1. The ferroptosis-susceptible cell state can either pre-exist in cells that arise from certain lineages or be acquired during cell-state transitions2-5. However, precisely how susceptibility to ferroptosis is dynamically regulated remains poorly understood. Here we use genome-wide CRISPR-Cas9 suppressor screens to identify the oxidative organelles peroxisomes as critical contributors to ferroptosis sensitivity in human renal and ovarian carcinoma cells. Using lipidomic profiling we show that peroxisomes contribute to ferroptosis by synthesizing polyunsaturated ether phospholipids (PUFA-ePLs), which act as substrates for lipid peroxidation that, in turn, results in the induction of ferroptosis. Carcinoma cells that are initially sensitive to ferroptosis can switch to a ferroptosis-resistant state in vivo in mice, which is associated with extensive downregulation of PUFA-ePLs. We further find that the pro-ferroptotic role of PUFA-ePLs can be extended beyond neoplastic cells to other cell types, including neurons and cardiomyocytes. Together, our work reveals roles for the peroxisome-ether-phospholipid axis in driving susceptibility to and evasion from ferroptosis, highlights PUFA-ePL as a distinct functional lipid class that is dynamically regulated during cell-state transitions, and suggests multiple regulatory nodes for therapeutic interventions in diseases that involve ferroptosis.


Subject(s)
Ethers/metabolism , Ferroptosis , Peroxisomes/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Ethers/chemistry , Female , Gene Editing , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lipid Peroxidation , Male , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurons/cytology , Neurons/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peroxisomes/genetics
2.
Cell Death Dis ; 15(4): 290, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658567

ABSTRACT

High-grade serous ovarian cancer (HGSOC) represents the most common and lethal subtype of ovarian cancer. Despite initial response to platinum-based standard therapy, patients commonly suffer from relapse that likely originates from drug-tolerant persister (DTP) cells. We generated isogenic clones of treatment-naïve and cisplatin-tolerant persister HGSOC cells. In addition, single-cell RNA sequencing of barcoded cells was performed in a xenograft model with HGSOC cell lines after platinum-based therapy. Published single-cell RNA-sequencing data from neo-adjuvant and non-treated HGSOC patients and patient data from TCGA were analyzed. DTP-derived cells exhibited morphological alterations and upregulation of epithelial-mesenchymal transition (EMT) markers. An aggressive subpopulation of DTP-derived cells showed high expression of the stress marker ATF3. Knockdown of ATF3 enhanced the sensitivity of aggressive DTP-derived cells to cisplatin-induced cell death, implying a role for ATF3 stress response in promoting a drug tolerant persister cell state. Furthermore, single cell lineage tracing to detect transcriptional changes in a HGSOC cell line-derived xenograft relapse model showed that cells derived from relapsed solid tumors express increased levels of EMT and multiple endoplasmic reticulum (ER) stress markers, including ATF3. Single cell RNA sequencing of epithelial cells from four HGSOC patients also identified a small cell population resembling DTP cells in all samples. Moreover, analysis of TCGA data from 259 HGSOC patients revealed a significant progression-free survival advantage for patients with low expression of the ATF3-associated partial EMT genes. These findings suggest that increased ATF3 expression together with partial EMT promote the development of aggressive DTP, and thereby relapse in HGSOC patients.


Subject(s)
Activating Transcription Factor 3 , Cisplatin , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Ovarian Neoplasms , Humans , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/genetics , Female , Cisplatin/pharmacology , Cisplatin/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Animals , Mice , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic/drug effects
3.
Nat Cell Biol ; 26(9): 1470-1481, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009641

ABSTRACT

Therapy resistance and metastasis, the most fatal steps in cancer, are often triggered by a (partial) activation of the epithelial-mesenchymal transition (EMT) programme. A mesenchymal phenotype predisposes to ferroptosis, a cell death pathway exerted by an iron and oxygen-radical-mediated peroxidation of phospholipids containing polyunsaturated fatty acids. We here show that various forms of EMT activation, including TGFß stimulation and acquired therapy resistance, increase ferroptosis susceptibility in cancer cells, which depends on the EMT transcription factor Zeb1. We demonstrate that Zeb1 increases the ratio of phospholipids containing pro-ferroptotic polyunsaturated fatty acids over cyto-protective monounsaturated fatty acids by modulating the differential expression of the underlying crucial enzymes stearoyl-Co-A desaturase 1 (SCD), fatty acid synthase (FASN), fatty acid desaturase 2 (FADS2), elongation of very long-chain fatty acid 5 (ELOVL5) and long-chain acyl-CoA synthetase 4 (ACSL4). Pharmacological inhibition of selected lipogenic enzymes (SCD and FADS2) allows the manipulation of ferroptosis sensitivity preferentially in high-Zeb1-expressing cancer cells. Our data are of potential translational relevance and suggest a combination of ferroptosis activators and SCD inhibitors for the treatment of aggressive cancers expressing high Zeb1.


Subject(s)
Epithelial-Mesenchymal Transition , Ferroptosis , Phospholipids , Stearoyl-CoA Desaturase , Zinc Finger E-box-Binding Homeobox 1 , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Humans , Cell Line, Tumor , Phospholipids/metabolism , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , Lipogenesis , Gene Expression Regulation, Neoplastic , Fatty Acid Desaturases/metabolism , Fatty Acid Desaturases/genetics , Animals , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/genetics , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Transforming Growth Factor beta/metabolism , Delta-5 Fatty Acid Desaturase , Drug Resistance, Neoplasm , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics
4.
bioRxiv ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38562716

ABSTRACT

Cancer cell fate has been widely ascribed to mutational changes within protein-coding genes associated with tumor suppressors and oncogenes. In contrast, the mechanisms through which the biophysical properties of membrane lipids influence cancer cell survival, dedifferentiation and metastasis have received little scrutiny. Here, we report that cancer cells endowed with a high metastatic ability and cancer stem cell-like traits employ ether lipids to maintain low membrane tension and high membrane fluidity. Using genetic approaches and lipid reconstitution assays, we show that these ether lipid-regulated biophysical properties permit non-clathrin-mediated iron endocytosis via CD44, leading directly to significant increases in intracellular redox-active iron and enhanced ferroptosis susceptibility. Using a combination of in vitro three-dimensional microvascular network systems and in vivo animal models, we show that loss of ether lipids also strongly attenuates extravasation, metastatic burden and cancer stemness. These findings illuminate a mechanism whereby ether lipids in carcinoma cells serve as key regulators of malignant progression while conferring a unique vulnerability that can be exploited for therapeutic intervention.

5.
Cell Rep ; 42(9): 113023, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37691145

ABSTRACT

Ferroptosis is a form of regulated cell death with roles in degenerative diseases and cancer. Excessive iron-catalyzed peroxidation of membrane phospholipids, especially those containing the polyunsaturated fatty acid arachidonic acid (AA), is central in driving ferroptosis. Here, we reveal that an understudied Golgi-resident scaffold protein, MMD, promotes susceptibility to ferroptosis in ovarian and renal carcinoma cells in an ACSL4- and MBOAT7-dependent manner. Mechanistically, MMD physically interacts with both ACSL4 and MBOAT7, two enzymes that catalyze sequential steps to incorporate AA in phosphatidylinositol (PI) lipids. Thus, MMD increases the flux of AA into PI, resulting in heightened cellular levels of AA-PI and other AA-containing phospholipid species. This molecular mechanism points to a pro-ferroptotic role for MBOAT7 and AA-PI, with potential therapeutic implications, and reveals that MMD is an important regulator of cellular lipid metabolism.


Subject(s)
Ferroptosis , Phosphatidylinositols , Cell Line , Fatty Acids, Unsaturated , Phosphatidylinositols/metabolism , Phospholipids/metabolism , Humans
6.
Nat Cell Biol ; 26(4): 516-518, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38589530
7.
Cancer Res ; 77(3): 790-801, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27940576

ABSTRACT

Despite the high incidence of oncogenic mutations in PIK3CA, the gene encoding the catalytic subunit of PI3K, PI3K inhibitors have yielded little clinical benefit for breast cancer patients. Recent epidemiologic studies have suggested a therapeutic benefit from aspirin intake in cancers harboring oncogenic PIK3CA Here, we show that mutant PIK3CA-expressing breast cancer cells have greater sensitivity to aspirin-mediated growth suppression than their wild-type counterparts. Aspirin decreased viability and anchorage-independent growth of mutant PIK3CA breast cancer cells independently of its effects on COX-2 and NF-κB. We ascribed the effects of aspirin to AMP-activated protein kinase (AMPK) activation, mTORC1 inhibition, and autophagy induction. In vivo, oncogenic PIK3CA-driven mouse mammary tumors treated daily with aspirin resulted in decreased tumor growth kinetics, whereas combination therapy of aspirin and a PI3K inhibitor further attenuated tumor growth. Our study supports the evaluation of aspirin and PI3K pathway inhibitors as a combination therapy for targeting breast cancer. Cancer Res; 77(3); 790-801. ©2016 AACR.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Female , Gene Knockdown Techniques , Humans , Immunoblotting , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred NOD , Mice, Transgenic , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
8.
Oncotarget ; 7(50): 81981-81994, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27626181

ABSTRACT

Long non-coding RNAs (lncRNAs) have been implicated in normal cellular homeostasis as well as pathophysiological conditions, including cancer. Here we performed global gene expression profiling of mammary epithelial cells transformed by oncogenic v-Src, and identified a large subset of uncharacterized lncRNAs potentially involved in breast cancer development. Specifically, our analysis revealed a novel lncRNA, LINC00520 that is upregulated upon ectopic expression of oncogenic v-Src, in a manner that is dependent on the transcription factor STAT3. Similarly, LINC00520 is also increased in mammary epithelial cells transformed by oncogenic PI3K and its expression is decreased upon knockdown of mutant PIK3CA. Additional expression profiling highlight that LINC00520 is elevated in a subset of human breast carcinomas, with preferential enrichment in the basal-like molecular subtype. ShRNA-mediated depletion of LINC00520 results in decreased cell migration and loss of invasive structures in 3D. RNA sequencing analysis uncovers several genes that are differentially expressed upon ectopic expression of LINC00520, a significant subset of which are also induced in v-Src-transformed MCF10A cells. Together, these findings characterize LINC00520 as a lncRNA that is regulated by oncogenic Src, PIK3CA and STAT3, and which may contribute to the molecular etiology of breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Class I Phosphatidylinositol 3-Kinases/metabolism , RNA, Long Noncoding/metabolism , STAT3 Transcription Factor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mammary Glands, Human/enzymology , Mammary Glands, Human/pathology , Mutation , Neoplasm Invasiveness , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , RNA Interference , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/genetics , Signal Transduction , Time Factors , Transfection , Up-Regulation
9.
Mol Oncol ; 9(6): 1140-54, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25735562

ABSTRACT

NFAT transcription factors are key regulators of gene expression in immune cells. In addition, NFAT1-induced genes play diverse roles in mediating the progression of various solid tumors. Here we show that NFAT1 induces the expression of the IL8 gene by binding to its promoter and leading to IL8 secretion. Thapsigargin stimulation of breast cancer cells induces IL8 expression in an NFAT-dependent manner. Moreover, we show that NFAT1-mediated IL8 production promotes the migration of primary human neutrophils in vitro and also promotes neutrophil infiltration in tumor xenografts. Furthermore, expression of active NFAT1 effectively suppresses the growth of nascent and established tumors by a non cell-autonomous mechanism. Evaluation of breast tumor tissue reveals that while the levels of NFAT1 are similar in tumor cells and normal breast epithelium, cells in the tumor stroma express higher levels of NFAT1 compared to normal stroma. Elevated levels of NFAT1 also correlate with increased neutrophil infiltrate in breast tumors. These data point to a mechanism by which NFAT1 orchestrates the communication between breast cancer cells and host neutrophils during breast cancer progression.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Interleukin-8/metabolism , NFATC Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Interleukin-8/genetics , Mice , Mice, Nude , NFATC Transcription Factors/genetics , Neoplasm Proteins/genetics , Neutrophils/pathology
SELECTION OF CITATIONS
SEARCH DETAIL