Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Mol Cell ; 78(6): 1192-1206.e10, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32470318

ABSTRACT

Tumor-derived extracellular vesicles are important mediators of cell-to-cell communication during tumorigenesis. Here, we demonstrated that hepatocellular carcinoma (HCC)-derived ectosomes remodel the tumor microenvironment to facilitate HCC progression in an ectosomal PKM2-dependent manner. HCC-derived ectosomal PKM2 induced not only metabolic reprogramming in monocytes but also STAT3 phosphorylation in the nucleus to upregulate differentiation-associated transcription factors, leading to monocyte-to-macrophage differentiation and tumor microenvironment remodeling. In HCC cells, sumoylation of PKM2 induced its plasma membrane targeting and subsequent ectosomal excretion via interactions with ARRDC1. The PKM2-ARRDC1 association in HCC was reinforced by macrophage-secreted cytokines/chemokines in a CCL1-CCR8 axis-dependent manner, further facilitating PKM2 excretion from HCC cells to form a feedforward regulatory loop for tumorigenesis. In the clinic, ectosomal PKM2 was clearly detected in the plasma of HCC patients. This study highlights a mechanism by which ectosomal PKM2 remodels the tumor microenvironment and reveals ectosomal PKM2 as a potential diagnostic marker for HCC.


Subject(s)
Carrier Proteins/metabolism , Cell-Derived Microparticles/metabolism , Membrane Proteins/metabolism , Thyroid Hormones/metabolism , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell-Derived Microparticles/genetics , Cell-Derived Microparticles/pathology , Chemokine CCL1/metabolism , Disease Progression , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Middle Aged , Monocytes/metabolism , Prognosis , STAT3 Transcription Factor/metabolism , Thyroid Hormones/genetics , Tumor Microenvironment , Thyroid Hormone-Binding Proteins
2.
Mol Cell ; 69(3): 480-492.e7, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395065

ABSTRACT

Fatty acid oxidation (FAO) is crucial for cells to overcome metabolic stress by providing ATP and NADPH. However, the mechanism by which FAO is regulated in tumors remains elusive. Here we show that Nur77 is required for the metabolic adaptation of melanoma cells by protecting FAO. Glucose deprivation activates ERK2 to phosphorylate and induce Nur77 translocation to the mitochondria, where Nur77 binds to TPß, a rate-limiting enzyme in FAO. Although TPß activity is normally inhibited by oxidation under glucose deprivation, the Nur77-TPß association results in Nur77 self-sacrifice to protect TPß from oxidation. FAO is therefore able to maintain NADPH and ATP levels and prevent ROS increase and cell death. The Nur77-TPß interaction further promotes melanoma metastasis by facilitating circulating melanoma cell survival. This study demonstrates a novel regulatory function of Nur77 with linkage of the FAO-NADPH-ROS pathway during metabolic stress, suggesting Nur77 as a potential therapeutic target in melanoma.


Subject(s)
Melanoma/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Animals , Cell Survival/physiology , Fatty Acids/metabolism , Glucose/metabolism , HEK293 Cells , Humans , Lipid Metabolism , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
3.
Proc Natl Acad Sci U S A ; 117(44): 27412-27422, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33087562

ABSTRACT

Nuclear receptor Nur77 participates in multiple metabolic regulations and plays paradoxical roles in tumorigeneses. Herein, we demonstrated that the knockout of Nur77 stimulated mammary tumor development in two mouse models, which would be reversed by a specific reexpression of Nur77 in mammary tissues. Mechanistically, Nur77 interacted and recruited corepressors, the SWI/SNF complex, to the promoters of CD36 and FABP4 to suppress their transcriptions, which hampered the fatty acid uptake, leading to the inhibition of cell proliferation. Peroxisome proliferator-activated receptor-γ (PPARγ) played an antagonistic role in this process through binding to Nur77 to facilitate ubiquitin ligase Trim13-mediated ubiquitination and degradation of Nur77. Cocrystallographic and functional analysis revealed that Csn-B, a Nur77-targeting compound, promoted the formation of Nur77 homodimer to prevent PPARγ binding by steric hindrance, thereby strengthening the Nur77's inhibitory role in breast cancer. Therefore, our study reveals a regulatory function of Nur77 in breast cancer via impeding fatty acid uptake.


Subject(s)
Breast Neoplasms/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , PPAR gamma/metabolism , Phenylacetates/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Proliferation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Fatty Acids/metabolism , Female , Humans , Kaplan-Meier Estimate , Lipid Metabolism/drug effects , Mammary Glands, Animal/pathology , Mice , Middle Aged , Nuclear Receptor Subfamily 4, Group A, Member 1/agonists , PPAR gamma/agonists , Primary Cell Culture , Prognosis , Proteolysis/drug effects , Tissue Array Analysis , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL