Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
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
4.
Nat Chem Biol ; 11(5): 339-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25822914

ABSTRACT

Sepsis, a hyperinflammatory response that can result in multiple organ dysfunctions, is a leading cause of mortality from infection. Here, we show that orphan nuclear receptor Nur77 (also known as TR3) can enhance resistance to lipopolysaccharide (LPS)-induced sepsis in mice by inhibiting NF-κB activity and suppressing aberrant cytokine production. Nur77 directly associates with p65 to block its binding to the κB element. However, this function of Nur77 is countered by the LPS-activated p38α phosphorylation of Nur77. Dampening the interaction between Nur77 and p38α would favor Nur77 suppression of the hyperinflammatory response. A compound, n-pentyl 2-[3,5-dihydroxy-2-(1-nonanoyl) phenyl]acetate, screened from a Nur77-biased library, blocked the Nur77-p38α interaction by targeting the ligand-binding domain of Nur77 and restored the suppression of the hyperinflammatory response through Nur77 inhibition of NF-κB. This study associates the nuclear receptor with immune homeostasis and implicates a new therapeutic strategy to treat hyperinflammatory responses by targeting a p38α substrate to modulate p38α-regulated functions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/prevention & control , Lipopolysaccharides/toxicity , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Phenylacetates/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , Animals , Diabetes Mellitus, Type 2/complications , Drug Evaluation, Preclinical , Homeostasis/drug effects , Inflammation/chemically induced , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Conformation , Sepsis/drug therapy , Sepsis/genetics , Transcription Factor RelA/antagonists & inhibitors
5.
Nat Chem Biol ; 10(2): 133-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316735

ABSTRACT

Autophagy is linked to cell death, yet the associated mechanisms are largely undercharacterized. We discovered that melanoma, which is generally resistant to drug-induced apoptosis, can undergo autophagic cell death with the participation of orphan nuclear receptor TR3. A sequence of molecular events leading to cellular demise is launched by a specific chemical compound, 1-(3,4,5-trihydroxyphenyl)nonan-1-one, newly acquired from screening a library of TR3-targeting compounds. The autophagic cascade comprises TR3 translocation to mitochondria through interaction with the mitochondrial outer membrane protein Nix, crossing into the mitochondrial inner membrane through Tom40 and Tom70 channel proteins, dissipation of mitochondrial membrane potential by the permeability transition pore complex ANT1-VDAC1 and induction of autophagy. This process leads to excessive mitochondria clearance and irreversible cell death. It implicates a new approach to melanoma therapy through activation of a mitochondrial signaling pathway that integrates a nuclear receptor with autophagy for cell death.


Subject(s)
Autophagy , Ketones/chemistry , Mitochondria/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Pyrogallol/analogs & derivatives , Signal Transduction , Animals , Cell Line, Tumor , Crystallography, X-Ray , Disease Models, Animal , Humans , Ketones/pharmacology , Melanoma/drug therapy , Membrane Proteins/metabolism , Mice , Protein Conformation , Proto-Oncogene Proteins/metabolism , Pyrogallol/chemistry , Pyrogallol/pharmacology , Tumor Suppressor Proteins/metabolism
6.
Nat Chem Biol ; 8(11): 897-904, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22983157

ABSTRACT

Liver kinase B1 (LKB1) has important roles in governing energy homeostasis by regulating the activity of the energy sensor kinase AMP-activated protein kinase (AMPK). The regulation of LKB1 function, however, is still poorly understood. Here we demonstrate that the orphan nuclear receptor Nur77 binds and sequesters LKB1 in the nucleus, thereby attenuating AMPK activation. This Nur77 function is antagonized by the chemical compound ethyl 2-[2,3,4-trimethoxy-6-(1-octanoyl)phenyl]acetate (TMPA), which interacts with Nur77 with high affinity and at specific sites. TMPA binding of Nur77 results in the release and shuttling of LKB1 to the cytoplasm to phosphorylate AMPKα. Moreover, TMPA effectively reduces blood glucose and alleviates insulin resistance in type II db/db and high-fat diet- and streptozotocin-induced diabetic mice but not in diabetic littermates with the Nur77 gene knocked out. This study attains a mechanistic understanding of the regulation of LKB1-AMPK axis and implicates Nur77 as a new and amenable target for the design and development of therapeutics to treat metabolic diseases.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phenylacetates/pharmacology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Blood Glucose/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Enzyme Activation/drug effects , HEK293 Cells , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Models, Molecular , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phenylacetates/chemistry , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Transport/drug effects , Streptozocin , Structure-Activity Relationship
7.
Gut ; 61(5): 714-24, 2012 May.
Article in English | MEDLINE | ID: mdl-21873734

ABSTRACT

AIMS: Wnt signalling is involved in cellular homeostasis and development. Dysregulation of the Wnt signalling pathway has been linked to colorectal cancer. The orphan nuclear receptor TR3 plays important roles in proliferation and apoptosis. In this study, we investigated how TR3 suppresses intestinal tumorigenesis by regulating Wnt signalling. METHODS: Intestinal polyps were quantified in Apc(min/+), Apc(min/+)/TR3(-/-) and Apc(min/+)/villin-TR3 mice. Wnt signalling activity was evaluated by assessing ß-galactosidase activity in a BAT-Gal reporter strain. The TR3 agonist cytosporone B was used to evaluate the role of TR3 in intestinal tumorigenesis. Crosstalk between TR3 and ß-catenin/TCF4 was analysed by molecular methods in colorectal cancer cells. The phosphorylation of TR3 by glycogen synthase kinase (GSK) 3ß and the correlation between GSK3ß activity and TR3 phosphorylation were evaluated in clinical samples and colorectal cancer cells. RESULTS: TR3 was found to significantly suppress Wnt signalling activity and the proliferation of intestinal epithelial cells. Apc(min/+)/TR3(-/-) mice developed more intestinal polyps than Apc(min/+)/TR3(+/+) mice, whereas either transgenic overexpression of TR3 in the intestine or treatment with cytosporone B in Apc(min/+) mice significantly decreased intestinal tumour number. Mechanistically, TR3 disrupted the association of ß-catenin and TCF4 on chromatin and facilitated the recruitment of transcriptional co-repressors to the promoters of Wnt signalling target genes. However, TR3 was phosphorylated by GSK3ß in most clinical colorectal cancers, which attenuated the inhibitory activity of TR3 towards Wnt signalling. CONCLUSIONS: TR3 is a negative regulator of Wnt signalling and thus significantly suppresses intestinal tumorigenesis in Apc(min/+) mice. This inhibitory effect of TR3 may be paradoxically overcome through phosphorylation by GSK3ß in clinical colorectal cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Intestinal Mucosa/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Colorectal Neoplasms/pathology , Down-Regulation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Intestinal Mucosa/pathology , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Transcription Factor 4 , beta Catenin/metabolism , beta-Galactosidase/metabolism
8.
Cell Res ; 33(12): 904-922, 2023 12.
Article in English | MEDLINE | ID: mdl-37460805

ABSTRACT

Pyroptosis is a type of regulated cell death executed by gasdermin family members. However, how gasdermin-mediated pyroptosis is negatively regulated remains unclear. Here, we demonstrate that mannose, a hexose, inhibits GSDME-mediated pyroptosis by activating AMP-activated protein kinase (AMPK). Mechanistically, mannose metabolism in the hexosamine biosynthetic pathway increases levels of the metabolite N-acetylglucosamine-6-phosphate (GlcNAc-6P), which binds AMPK to facilitate AMPK phosphorylation by LKB1. Activated AMPK then phosphorylates GSDME at Thr6, which leads to blockade of caspase-3-induced GSDME cleavage, thereby repressing pyroptosis. The regulatory role of AMPK-mediated GSDME phosphorylation was further confirmed in AMPK knockout and GSDMET6E or GSDMET6A knock-in mice. In mouse primary cancer models, mannose administration suppressed pyroptosis in small intestine and kidney to alleviate cisplatin- or oxaliplatin-induced tissue toxicity without impairing antitumor effects. The protective effect of mannose was also verified in a small group of patients with gastrointestinal cancer who received normal chemotherapy. Our study reveals a novel mechanism whereby mannose antagonizes GSDME-mediated pyroptosis through GlcNAc-6P-mediated activation of AMPK, and suggests the utility of mannose supplementation in alleviating chemotherapy-induced side effects in clinic applications.


Subject(s)
Mannose , Pyroptosis , Humans , Animals , Mice , Mannose/pharmacology , AMP-Activated Protein Kinases , Gasdermins
9.
Cell Chem Biol ; 30(3): 261-277.e8, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36889311

ABSTRACT

Pulmonary fibrosis is a typical sequela of coronavirus disease 2019 (COVID-19), which is linked with a poor prognosis for COVID-19 patients. However, the underlying mechanism of pulmonary fibrosis induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is unclear. Here, we demonstrated that the nucleocapsid (N) protein of SARS-CoV-2 induced pulmonary fibrosis by activating pulmonary fibroblasts. N protein interacted with the transforming growth factor ß receptor I (TßRI), to disrupt the interaction of TßRI-FK506 Binding Protein12 (FKBP12), which led to activation of TßRI to phosphorylate Smad3 and boost expression of pro-fibrotic genes and secretion of cytokines to promote pulmonary fibrosis. Furthermore, we identified a compound, RMY-205, that bound to Smad3 to disrupt TßRI-induced Smad3 activation. The therapeutic potential of RMY-205 was strengthened in mouse models of N protein-induced pulmonary fibrosis. This study highlights a signaling pathway of pulmonary fibrosis induced by N protein and demonstrates a novel therapeutic strategy for treating pulmonary fibrosis by a compound targeting Smad3.


Subject(s)
COVID-19 , Pulmonary Fibrosis , Animals , Mice , COVID-19/complications , Fibrosis , Nucleocapsid Proteins/therapeutic use , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/drug therapy , SARS-CoV-2
10.
Carcinogenesis ; 33(2): 301-11, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22159226

ABSTRACT

Cisplatin is a widely used antitumor agent that induces aggressive cancer cell death via triggering cellular proteins involved in apoptosis. Here, we demonstrate that cisplatin effectively induces orphan nuclear receptor TR3 phosphorylation by activating Chk2 kinase activity and promoting cross talk between these two proteins, thereby contributing to the repression of intestinal tumorigenesis via apoptosis. Mechanistic analysis has demonstrated that Chk2-induced phosphorylation enables TR3 to bind to its response elements on the promoters of the BRE and RNF-7 genes, leading to the negative regulation of these two anti-apoptotic genes. Furthermore, the induction of apoptosis by cisplatin is mediated by TR3, and knockdown of TR3 reduces cisplatin-induced apoptosis in colon cancer cells by 27%. The role of TR3 in cisplatin chemotherapy is further clarified in mouse models. In Apc(min/+) mice, cisplatin inhibits intestinal tumorigenesis by 70% in a TR3 phosphorylation-dependent manner; however, the loss of TR3 function in Apc(min/+)/TR3(-/-) mice leads to the failure of cisplatin-induced repression of tumorigenesis. Consistently, xenografts derived from TR3 knockdown colon cancer cells are insensitive to cisplatin treatment, whereas a significant curative effect (50% inhibition) is observed in xenografts with functional TR3. Taken together, our study reveals a novel cross talk between Chk2 and TR3 and sheds light on the mechanism of cisplatin-induced apoptosis through TR3. Therefore, TR3 may be a new target of cisplatin for colon cancer therapy.


Subject(s)
Apoptosis/drug effects , Cell Transformation, Neoplastic/drug effects , Cisplatin/pharmacology , Intestinal Neoplasms/prevention & control , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Checkpoint Kinase 2 , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Gene Knockdown Techniques/methods , HEK293 Cells , Humans , Intestinal Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transplantation, Heterologous/methods
11.
Nat Metab ; 4(10): 1306-1321, 2022 10.
Article in English | MEDLINE | ID: mdl-36192599

ABSTRACT

Extracellular vesicles play crucial roles in intercellular communication in the tumor microenvironment. Here we demonstrate that in hepatic fibrosis, TGF-ß stimulates the palmitoylation of hexokinase 1 (HK1) in hepatic stellate cells (HSCs), which facilitates the secretion of HK1 via large extracellular vesicles in a TSG101-dependent manner. The large extracellular vesicle HK1 is hijacked by hepatocellular carcinoma (HCC) cells, leading to accelerated glycolysis and HCC progression. In HSCs, the nuclear receptor Nur77 transcriptionally activates the expression of depalmitoylase ABHD17B to inhibit HK1 palmitoylation, consequently attenuating HK1 release. However, TGF-ß-activated Akt functionally represses Nur77 by inducing Nur77 phosphorylation and degradation. We identify the small molecule PDNPA that binds Nur77 to generate steric hindrance to block Akt targeting, thereby disrupting Akt-mediated Nur77 degradation and preserving Nur77 inhibition of HK1 release. Together, this study demonstrates an overlooked function of HK1 in HCC upon its release from HSCs and highlights PDNPA as a candidate compound for inhibiting HCC progression.


Subject(s)
Carcinoma, Hepatocellular , Extracellular Vesicles , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Hepatic Stellate Cells/metabolism , Hexokinase/metabolism , Liver Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Proliferation , Cell Line, Tumor , Extracellular Vesicles/metabolism , Transforming Growth Factor beta/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Microenvironment
12.
Nucleic Acids Res ; 37(3): 832-48, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19095693

ABSTRACT

PRMT1, an arginine methyltransferase, plays an important role in numerous cellular processes. In this study, we demonstrate a feedback regulatory loop between PRMT1 and the orphan receptor TR3. Unlike another orphan receptor HNF4, TR3 is not methylated by PRMT1 although they physically interact with each other. By delaying the TR3 protein degradation, PRMT1 binding leads to the elevation of TR3 cellular protein level, thereby enhances the DNA binding and transactivation activity of TR3 in a non-methyltransferase manner. Another coactivator SRC-2 acts synergistically with PRMT1 to regulate TR3 functions. In turn, TR3 binding to the catalytic domain of PRMT1 causes an inhibition of the PRMT1 methyltransferase activity. This repression results in the functional changes in some of PRMT1 substrates, including STAT3 and Sam68. The negative regulation of PRMT1 by TR3 was further confirmed in both TR3-knockdown cells and TR3-knockout mice with the use of an agonist for TR3. Taken together, our study not only identifies a regulatory role of PRMT1, independent on methyltransferase activity, in TR3 transactivation, but also characterizes a novel function of TR3 in the repression of PRMT1 methyltransferase activity.


Subject(s)
DNA-Binding Proteins/metabolism , Feedback, Physiological , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Steroid/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , DNA-Binding Proteins/agonists , Humans , Mice , Mice, Knockout , Nuclear Receptor Coactivator 2/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Receptors, Steroid/agonists , Repressor Proteins/antagonists & inhibitors
13.
Cancer Lett ; 516: 48-56, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34082025

ABSTRACT

Extracellular vesicles (EVs) have gained significant attention in recent decades as major mediators of intercellular communication that are involved in various essential physiological and pathological processes. They are secreted by almost all cell types and carry bioactive materials, such as proteins, lipids and nucleic acids, that can be transmitted from host cells to recipient cells, thereby eliciting phenotypic and functional alterations in the recipient cells. Recent evidence shows that EVs play essential roles in remodeling the tumor immune microenvironment (TIME). EVs derived from tumor cells and immune cells mediate mutual communication at proximal and distal sites, which determines tumor fate and antitumor therapeutic effectiveness. In this review, the current understanding of EVs and their roles in remodeling the TIME and modulating tumor-specific immunity are summarized. We mainly discuss the mutual regulation between tumor cells and tumor-infiltrating immune cells through the delivery of EVs in the TIME. We also describe the limitations of current studies and discuss directions for further research.


Subject(s)
Extracellular Vesicles/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Humans , Lymphocytes, Tumor-Infiltrating/immunology
14.
Cell Res ; 31(9): 980-997, 2021 09.
Article in English | MEDLINE | ID: mdl-34012073

ABSTRACT

Pyroptosis is a form of regulated cell death mediated by gasdermin family members, among which the function of GSDMC has not been clearly described. Herein, we demonstrate that the metabolite α-ketoglutarate (α-KG) induces pyroptosis through caspase-8-mediated cleavage of GSDMC. Treatment with DM-αKG, a cell-permeable derivative of α-KG, elevates ROS levels, which leads to oxidation of the plasma membrane-localized death receptor DR6. Oxidation of DR6 triggers its endocytosis, and then recruits both pro-caspase-8 and GSDMC to a DR6 receptosome through protein-protein interactions. The DR6 receptosome herein provides a platform for the cleavage of GSDMC by active caspase-8, thereby leading to pyroptosis. Moreover, this α-KG-induced pyroptosis could inhibit tumor growth and metastasis in mouse models. Interestingly, the efficiency of α-KG in inducing pyroptosis relies on an acidic environment in which α-KG is reduced by MDH1 and converted to L-2HG that further boosts ROS levels. Treatment with lactic acid, the end product of glycolysis, builds an improved acidic environment to facilitate more production of L-2HG, which makes the originally pyroptosis-resistant cancer cells more susceptible to α-KG-induced pyroptosis. This study not only illustrates a pyroptotic pathway linked with metabolites but also identifies an unreported principal axis extending from ROS-initiated DR6 endocytosis to caspase-8-mediated cleavage of GSDMC for potential clinical application in tumor therapy.


Subject(s)
Caspase 8 , DNA-Binding Proteins , Neoplasms , Pyroptosis , Receptors, Tumor Necrosis Factor , Animals , Caspase 1/metabolism , Ketoglutaric Acids , Mice , Receptors, Death Domain
15.
Oncogene ; 39(11): 2408-2423, 2020 03.
Article in English | MEDLINE | ID: mdl-31959898

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality worldwide. Orphan nuclear receptor Nur77, which is low expressed in HCC, functions as a tumor suppressor to suppress HCC. However, the detailed mechanism is still not well understood. Here, we demonstrate that Nur77 could inhibit HCC development via transcriptional activation of the lncRNA WAP four-disulfide core domain 21 pseudogene (WFDC21P). Nur77 binds to its response elements on the WFDC21P promoter to directly induce WFDC21P transcription, which inhibits HCC cell proliferation, tumor growth, and tumor metastasis both in vitro and in vivo. In clinical HCC samples, WFDC21P expression positively correlated with that of Nur77, and the loss of WFDC21P is associated with worse prognosis. Mechanistically, WFDC21P could inhibit glycolysis by simultaneously interacting with PFKP and PKM2, two key enzymes in glycolysis. These interactions not only abrogate the tetramer formation of PFKP to impede its catalytic activity but also prevent the nuclear translocation of PKM2 to suppress its function as a transcriptional coactivator. Cytosporone-B (Csn-B), an agonist for Nur77, could stimulate WFDC21P expression and suppress HCC in a WFDC21P-dependent manner. Therefore, our study reveals a new HCC suppressor and connects the glycolytic remodeling of HCC with the Nur77-WFDC21P-PFKP/PKM2 axis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , RNA, Long Noncoding/metabolism , Animals , Carcinogenesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Growth Processes , Cell Line, Tumor , Glycolysis , Hep G2 Cells , Heterografts , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Receptor Subfamily 4, Group A, Member 1/agonists , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phenylacetates/pharmacology , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , Transcriptional Activation , Up-Regulation
16.
Cell Rep ; 33(3): 108284, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086070

ABSTRACT

The Hippo signaling pathway maintains organ size and tissue homeostasis via orchestration of cell proliferation and apoptosis. How this pathway triggers cell apoptosis remains largely unexplored. Here, we identify NR4A1 as a target of the Hippo pathway that mediates the pro-apoptotic and anti-tumor effects of the Hippo pathway whereby YAP regulates the transcription, phosphorylation, and mitochondrial localization of NR4A1. NR4A1, in turn, functions as a feedback inhibitor of YAP to promote its degradation, thereby inhibiting the function of YAP during liver regeneration and tumorigenesis. Our studies elucidate a regulatory loop between NR4A1 and YAP to coordinate Hippo signaling activity during liver regeneration and tumorigenesis and highlight NR4A1 as a marker of Hippo signaling, as well as a therapeutic target for hepatocellular carcinoma.


Subject(s)
Cell Cycle Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/physiology , Carcinogenesis , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Female , Homeostasis/physiology , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphorylation , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription Factors/genetics , YAP-Signaling Proteins
17.
Nucleic Acids Res ; 35(21): 7348-59, 2007.
Article in English | MEDLINE | ID: mdl-17962304

ABSTRACT

p300 regulates the transcriptional activity of a variety of transcription factors by forming an activation complex and/or promoting histone acetylation. Here, we show a unique characteristic of orphan receptor TR3 in negatively regulating the function of p300. TR3 was found to interact with p300 and inhibited the acetylation of transcription factors induced by p300, resulting in the repression of their transcriptional activity. Further analysis revealed that both a conserved transcriptional adapter motif (TRAM) in p300 and a specific sequence FLELFIL in TR3 were critical for their interaction. TR3 binding completely covered the histone acetyltransferase (HAT) domain of p300 and resulted in suppression of the HAT activity, as the p300-induced histone H3 acetylation and transcription were inhibited with the presence TR3. Furthermore, an agonist of TR3, a natural octaketide isolated from Dothiorella sp. HTF3 of an endophytical fungus, was shown to be a potent compound for inhibiting p300 HAT activity (IC(50) = 1.5 microg/ml) in vivo. More importantly, this agonist could repress the transcriptional activity of transcription factors, and proliferation of cancer cells. Taken together, our results not only delineate a novel transcriptional repressor function for TR3, but also reveal its modulation on p300 HAT activity as the underlying mechanism.


Subject(s)
Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Repressor Proteins/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Acetylation , Amino Acid Sequence , Binding Sites , Cell Line , Down-Regulation , Gene Expression Regulation , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1 , Receptors, Steroid/agonists , Receptors, Steroid/chemistry , Receptors, Thyroid Hormone/agonists , Receptors, Thyroid Hormone/chemistry , Repressor Proteins/agonists , Repressor Proteins/chemistry , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/metabolism
18.
Carcinogenesis ; 29(11): 2078-88, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18713840

ABSTRACT

Acutely transforming retrovirus AKT8 in rodent T cell lymphoma (Akt) phosphorylates and regulates the function of many cellular proteins involved in processes such as metabolism, apoptosis and proliferation. However, the precise mechanisms by which Akt promotes cell survival and inhibits apoptosis have been characterized in part only. TR3, an orphan receptor, functions as a transcription factor that can both positively or negatively regulate gene expression. We have reported previously that the translocation of TR3 from the nucleus to the mitochondria can elicit a proapoptotic effect in gastric cancer cells. In our present study, we demonstrate that Akt phosphorylates cytoplasmic TR3 through its physical interaction with the N-terminus of TR3. When coexpressed with Akt, TR3 mitochondrial targeting was blocked and this protein adopted a diffuse expression pattern in the cytoplasm. Moreover, Akt displayed an ability to disrupt the interaction of TR3 with Bcl-2, which is thought to be a critical requirement for mitochondrial TR3 to elicit apoptosis. Consistently, insulin was also found to induce the phosphorylation of TR3 and abolish 12-O-tetradecanoylphorbol-13-acetate-induced mitochondrial localization, which was dependent upon the activation of the phophatidylinositol-3-OH-kinase-Akt signaling pathway. Taken together, our current data demonstrate a unique role for Akt in inhibiting TR3 functions that are not related to transcriptional activity but that correlate with the regulation of its mitochondrial association. This may represent a novel signal pathway by which Akt exerts its antiapoptotic effects in gastric cancer cells, i.e. by regulating the phosphorylation and redistribution of orphan receptors.


Subject(s)
Mitochondria/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Apoptosis , Base Sequence , Binding Sites , Blotting, Western , Cell Line, Tumor , Cytochromes c/metabolism , Cytoplasm/metabolism , DNA Primers , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Nuclear Receptor Subfamily 4, Group A, Member 1 , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Steroid/chemistry , Receptors, Thyroid Hormone/chemistry
19.
Mol Endocrinol ; 21(12): 2877-89, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761950

ABSTRACT

Acetylation modification regulates the functions of histone and nonhistone proteins, including transcriptional activity, protein interaction, and subcellular localization. Although many nuclear receptors have been shown to be modified by acetylation, whether retinoid X receptors (RXRs) are acetylated and how the acetylation is regulated remains unknown. Here, we provide the first evidence of RXRalpha acetylation by p300 on lysine 145. Acetylation of RXRalpha by p300 facilitated its DNA binding and subsequently increased its transcriptional activity. Furthermore, we discovered that TR3, an orphan receptor, exerted a negative regulation on p300-induced RXRalpha acetylation. TR3 significantly reduced the p300-induced RXRalpha acetylation and transcriptional activity, and such inhibition required the interaction of TR3 with RXRalpha. Binding of TR3 to RXRalpha resulted in the sequestration of RXRalpha from p300. 9-cis retinoic acid, a ligand for RXRalpha, enhanced the association of RXRalpha with TR3, rather than acetylation of RXRalpha by p300. Biological function analysis revealed that the mitogenic activity of RXRalpha stimulated by p300 was acetylation dependent and could be repressed by TR3. Upon the treatment of 9-cis retinoic acid, RXRalpha was translocated with TR3 from the nucleus to the mitochondria, and apoptosis was induced. Taken together, our data demonstrate the distinct regulatory mechanisms of p300 and TR3 on RXRalpha acetylation and reveal a previously unrecognized role for orphan receptor in the transcriptional control of retinoid receptors.


Subject(s)
E1A-Associated p300 Protein/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Retinoid X Receptors/metabolism , Acetylation , Alitretinoin , Apoptosis , Cell Line , DNA/metabolism , Humans , Lysine/genetics , Lysine/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Protein Binding , Protein Transport , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Retinoid X Receptors/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Tretinoin/pharmacology
20.
Cell Res ; 28(12): 1171-1185, 2018 12.
Article in English | MEDLINE | ID: mdl-30287942

ABSTRACT

Iron has been shown to trigger oxidative stress by elevating reactive oxygen species (ROS) and to participate in different modes of cell death, such as ferroptosis, apoptosis and necroptosis. However, whether iron-elevated ROS is also linked to pyroptosis has not been reported. Here, we demonstrate that iron-activated ROS can induce pyroptosis via a Tom20-Bax-caspase-GSDME pathway. In melanoma cells, iron enhanced ROS signaling initiated by CCCP, causing the oxidation and oligomerization of the mitochondrial outer membrane protein Tom20. Bax is recruited to mitochondria by oxidized Tom20, which facilitates cytochrome c release to cytosol to activate caspase-3, eventually triggering pyroptotic death by inducing GSDME cleavage. Therefore, ROS acts as a causative factor and Tom20 senses ROS signaling for iron-driven pyroptotic death of melanoma cells. Since iron activates ROS for GSDME-dependent pyroptosis induction and melanoma cells specifically express a high level of GSDME, iron may be a potential candidate for melanoma therapy. Based on the functional mechanism of iron shown above, we further demonstrate that iron supplementation at a dosage used in iron-deficient patients is sufficient to maximize the anti-tumor effect of clinical ROS-inducing drugs to inhibit xenograft tumor growth and metastasis of melanoma cells through GSDME-dependent pyroptosis. Moreover, no obvious side effects are observed in the normal tissues and organs of mice during the combined treatment of clinical drugs and iron. This study not only identifies iron as a sensitizer amplifying ROS signaling to drive pyroptosis, but also implicates a novel iron-based intervention strategy for melanoma therapy.


Subject(s)
Iron/pharmacology , Melanoma/metabolism , Membrane Transport Proteins/metabolism , Mitochondria , Pyroptosis/drug effects , Receptors, Cell Surface/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , HEK293 Cells , Humans , Melanoma/drug therapy , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL