Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 563
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(7): 835-851, 2019 07.
Article in English | MEDLINE | ID: mdl-31160797

ABSTRACT

How tumor cells genetically lose antigenicity and evade immune checkpoints remains largely elusive. We report that tissue-specific expression of the human long noncoding RNA LINK-A in mouse mammary glands initiates metastatic mammary gland tumors, which phenotypically resemble human triple-negative breast cancer (TNBC). LINK-A expression facilitated crosstalk between phosphatidylinositol-(3,4,5)-trisphosphate and inhibitory G-protein-coupled receptor (GPCR) pathways, attenuating protein kinase A-mediated phosphorylation of the E3 ubiquitin ligase TRIM71. Consequently, LINK-A expression enhanced K48-polyubiquitination-mediated degradation of the antigen peptide-loading complex (PLC) and intrinsic tumor suppressors Rb and p53. Treatment with LINK-A locked nucleic acids or GPCR antagonists stabilized the PLC components, Rb and p53, and sensitized mammary gland tumors to immune checkpoint blockers. Patients with programmed ccll death protein-1(PD-1) blockade-resistant TNBC exhibited elevated LINK-A levels and downregulated PLC components. Hence we demonstrate lncRNA-dependent downregulation of antigenicity and intrinsic tumor suppression, which provides the basis for developing combinational immunotherapy treatment regimens and early TNBC prevention.


Subject(s)
Antigen Presentation/immunology , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/immunology , Oncogenes , RNA, Long Noncoding/genetics , Tumor Escape/genetics , Tumor Escape/immunology , Adenoma/genetics , Adenoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Disease Progression , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Receptors, G-Protein-Coupled/antagonists & inhibitors , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
2.
Cell ; 161(2): 240-54, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25860607

ABSTRACT

In vitro modeling of human disease has recently become feasible with induced pluripotent stem cell (iPSC) technology. Here, we established patient-derived iPSCs from a Li-Fraumeni syndrome (LFS) family and investigated the role of mutant p53 in the development of osteosarcoma (OS). LFS iPSC-derived osteoblasts (OBs) recapitulated OS features including defective osteoblastic differentiation as well as tumorigenic ability. Systematic analyses revealed that the expression of genes enriched in LFS-derived OBs strongly correlated with decreased time to tumor recurrence and poor patient survival. Furthermore, LFS OBs exhibited impaired upregulation of the imprinted gene H19 during osteogenesis. Restoration of H19 expression in LFS OBs facilitated osteoblastic differentiation and repressed tumorigenic potential. By integrating human imprinted gene network (IGN) into functional genomic analyses, we found that H19 mediates suppression of LFS-associated OS through the IGN component DECORIN (DCN). In summary, these findings demonstrate the feasibility of studying inherited human cancer syndromes with iPSCs.


Subject(s)
Gene Regulatory Networks , Induced Pluripotent Stem Cells/cytology , Li-Fraumeni Syndrome/complications , Osteosarcoma/etiology , Adolescent , Adult , Animals , Child , Decorin/metabolism , Female , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/pathology , Male , Mesenchymal Stem Cells/metabolism , Mice , Models, Biological , Neoplasm Transplantation , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Long Noncoding/metabolism , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
3.
Cell ; 159(5): 1110-1125, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416949

ABSTRACT

lncRNAs are known to regulate a number of different developmental and tumorigenic processes. Here, we report a role for lncRNA BCAR4 in breast cancer metastasis that is mediated by chemokine-induced binding of BCAR4 to two transcription factors with extended regulatory consequences. BCAR4 binding of SNIP1 and PNUTS in response to CCL21 releases the SNIP1's inhibition of p300-dependent histone acetylation, which in turn enables the BCAR4-recruited PNUTS to bind H3K18ac and relieve inhibition of RNA Pol II via activation of the PP1 phosphatase. This mechanism activates a noncanonical Hedgehog/GLI2 transcriptional program that promotes cell migration. BCAR4 expression correlates with advanced breast cancers, and therapeutic delivery of locked nucleic acids (LNAs) targeting BCAR4 strongly suppresses breast cancer metastasis in mouse models. The findings reveal a disease-relevant lncRNA mechanism consisting of both direct coordinated protein recruitment and indirect regulation of transcription factors.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Metastasis , RNA, Long Noncoding/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kruppel-Like Transcription Factors/genetics , Mice , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Transcriptional Activation , Zinc Finger Protein Gli2 , p300-CBP Transcription Factors/metabolism
4.
Mol Cell ; 81(22): 4579-4590, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34562371

ABSTRACT

Canonically, gasdermin D (GSDMD) cleavage by caspase-1 through inflammasome signaling triggers immune cell pyroptosis (ICP) as a host defense against pathogen infection. However, cancer cell pyroptosis (CCP) was recently discovered to be activated by distinct molecular mechanisms in which GSDMB, GSDMC, and GSDME, rather than GSDMD, are the executioners. Moreover, instead of inflammatory caspases, apoptotic caspases and granzymes are required for gasdermin protein cleavage to induce CCP. Sufficient accumulation of protease-cleaved gasdermin proteins is the prerequisite for CCP. Inflammation induced by ICP or CCP results in diametrically opposite effects on antitumor immunity because of the differential duration and released cellular contents, leading to contrary effects on therapeutic outcomes. Here, we focus on the distinct mechanisms of ICP and CCP and discuss the roles of ICP and CCP in inflammation and antitumor immunity, representing actionable targets.


Subject(s)
Antineoplastic Agents/pharmacology , Inflammation , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/immunology , Neoplasms/therapy , Phosphate-Binding Proteins/metabolism , Pyroptosis , Animals , Apoptosis , Caspases/metabolism , Cell Line, Tumor , Disease Progression , Humans , Immune System , Inflammasomes , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/metabolism , Mice , Neoplasms/metabolism , Protein Binding , Proteolysis , Signal Transduction
5.
Mol Cell ; 81(11): 2317-2331.e6, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33909988

ABSTRACT

Aberrant energy status contributes to multiple metabolic diseases, including obesity, diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4 immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on Ser283, thereby disrupting its interaction with CMTM4 and subsequently triggering PD-L1 degradation. In addition, AMPK phosphorylates EZH2, which disrupts PRC2 function, leading to enhanced IFNs and antigen presentation gene expression. Through these mechanisms, AMPK agonists or ketogenic diets enhance the efficacy of anti-CTLA-4 immunotherapy and improve the overall survival rate in syngeneic mouse tumor models. Our findings reveal a pivotal role for AMPK in regulating the immune response to immune-checkpoint blockade and advocate for combining ketogenic diets or AMPK agonists with anti-CTLA4 immunotherapy to combat cancer.


Subject(s)
AMP-Activated Protein Kinases/genetics , B7-H1 Antigen/genetics , Breast Neoplasms/genetics , CTLA-4 Antigen/genetics , Colorectal Neoplasms/genetics , Immune Checkpoint Inhibitors , AMP-Activated Protein Kinases/immunology , Allografts , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , B7-H1 Antigen/immunology , Biphenyl Compounds/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/therapy , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Diet, Ketogenic/methods , Energy Metabolism/drug effects , Energy Metabolism/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Nude , Pyrones/pharmacology , Signal Transduction , Survival Analysis , Thiophenes/pharmacology
6.
Trends Biochem Sci ; 2024 May 06.
Article in English | MEDLINE | ID: mdl-38714376

ABSTRACT

Protein lipoylation, a crucial post-translational modification (PTM), plays a pivotal role in mitochondrial function and emerges as a key player in cell death through cuproptosis. This novel copper-driven cell death pathway is activated by excessive copper ions binding to lipoylated mitochondrial proteins, disrupting energy production and causing lethal protein aggregation and cell death. The intricate relationship among protein lipoylation, cellular energy metabolism, and cuproptosis offers a promising avenue for regulating essential cellular functions. This review focuses on the mechanisms of lipoylation and its significant impact on cell metabolism and cuproptosis, emphasizing the key genes involved and their implications for human diseases. It offers valuable insights into targeting dysregulated cellular metabolism for therapeutic purposes.

7.
Cell ; 154(3): 556-68, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911321

ABSTRACT

Skp2 E3 ligase is overexpressed in numerous human cancers and plays a critical role in cell-cycle progression, senescence, metabolism, cancer progression, and metastasis. In the present study, we identified a specific Skp2 inhibitor using high-throughput in silico screening of large and diverse chemical libraries. This Skp2 inhibitor selectively suppresses Skp2 E3 ligase activity, but not activity of other SCF complexes. It also phenocopies the effects observed upon genetic Skp2 deficiency, such as suppressing survival and Akt-mediated glycolysis and triggering p53-independent cellular senescence. Strikingly, we discovered a critical function of Skp2 in positively regulating cancer stem cell populations and self-renewal ability through genetic and pharmacological approaches. Notably, Skp2 inhibitor exhibits potent antitumor activities in multiple animal models and cooperates with chemotherapeutic agents to reduce cancer cell survival. Our study thus provides pharmacological evidence that Skp2 is a promising target for restricting cancer stem cell and cancer progression.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Neoplasms/enzymology , Neoplastic Stem Cells/drug effects , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Disease Models, Animal , Drug Screening Assays, Antitumor , Genes, p53 , Glycolysis/drug effects , Humans , Mice , Mice, Nude , Models, Molecular , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/metabolism , Small Molecule Libraries , Structure-Activity Relationship , Transplantation, Heterologous , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
8.
Mol Cell ; 80(4): 633-647.e7, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33217317

ABSTRACT

N6-methyladenosine (m6A) is the most abundant mRNA modification and is installed by the METTL3-METTL14-WTAP methyltransferase complex. Although the importance of m6A methylation in mRNA metabolism has been well documented recently, regulation of the m6A machinery remains obscure. Through a genome-wide CRISPR screen, we identify the ERK pathway and USP5 as positive regulators of the m6A deposition. We find that ERK phosphorylates METTL3 at S43/S50/S525 and WTAP at S306/S341, followed by deubiquitination by USP5, resulting in stabilization of the m6A methyltransferase complex. Lack of METTL3/WTAP phosphorylation reduces decay of m6A-labeled pluripotent factor transcripts and traps mouse embryonic stem cells in the pluripotent state. The same phosphorylation can also be found in ERK-activated human cancer cells and contribute to tumorigenesis. Our study reveals an unrecognized function of ERK in regulating m6A methylation.


Subject(s)
Adenine/analogs & derivatives , Carcinogenesis/pathology , Endopeptidases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Melanoma/pathology , Methyltransferases/chemistry , Adenine/chemistry , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endopeptidases/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Methyltransferases/physiology , Mice , Mice, Knockout , Phosphorylation , Protein Stability , RNA Processing, Post-Transcriptional
9.
Immunity ; 49(3): 504-514.e4, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30231984

ABSTRACT

The adaptor protein CARD9 links detection of fungi by surface receptors to the activation of the NF-κB pathway. Mice deficient in CARD9 exhibit dysbiosis and are more susceptible to colitis. Here we examined the impact of Card9 deficiency in the development of colitis-associated colon cancer (CAC). Treatment of Card9-/- mice with AOM-DSS resulted in increased tumor loads as compared to WT mice and in the accumulation of myeloid-derived suppressor cells (MDSCs) in tumor tissue. The impaired fungicidal functions of Card9-/- macrophages led to increased fungal loads and variation in the overall composition of the intestinal mycobiota, with a notable increase in C. tropicalis. Bone marrow cells incubated with C. tropicalis exhibited MDSC features and suppressive functions. Fluconazole treatment suppressed CAC in Card9-/- mice and was associated with decreased MDSC accumulation. The frequency of MDSCs in tumor tissues of colon cancer patients correlated positively with fungal burden, pointing to the relevance of this regulatory axis in human disease.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Colitis/immunology , Colonic Neoplasms/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Myeloid-Derived Suppressor Cells/physiology , Animals , CARD Signaling Adaptor Proteins/genetics , Cell Proliferation , Cells, Cultured , Coculture Techniques , Colitis/chemically induced , Colitis/genetics , Colonic Neoplasms/genetics , Dysbiosis/genetics , Humans , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Derived Suppressor Cells/microbiology , Promoter Regions, Genetic/genetics
10.
Cell ; 149(5): 1098-111, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632973

ABSTRACT

Akt kinase plays a central role in cell growth, metabolism, and tumorigenesis. The TRAF6 E3 ligase orchestrates IGF-1-mediated Akt ubiquitination and activation. Here, we show that Akt ubiquitination is also induced by activation of ErbB receptors; unexpectedly, and in contrast to IGF-1 induced activation, the Skp2 SCF complex, not TRAF6, is a critical E3 ligase for ErbB-receptor-mediated Akt ubiquitination and membrane recruitment in response to EGF. Skp2 deficiency impairs Akt activation, Glut1 expression, glucose uptake and glycolysis, and breast cancer progression in various tumor models. Moreover, Skp2 overexpression correlates with Akt activation and breast cancer metastasis and serves as a marker for poor prognosis in Her2-positive patients. Finally, Skp2 silencing sensitizes Her2-overexpressing tumors to Herceptin treatment. Our study suggests that distinct E3 ligases are utilized by diverse growth factors for Akt activation and that targeting glycolysis sensitizes Her2-positive tumors to Herceptin treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/drug therapy , Cell Transformation, Neoplastic , F-Box Proteins/metabolism , Glycolysis , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Humans , Mice , Receptor, ErbB-2/metabolism , S-Phase Kinase-Associated Proteins/genetics , Trastuzumab , Ubiquitination
11.
Mol Cell ; 76(3): 359-370, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31668929

ABSTRACT

The engagement of programmed cell death protein 1 (PD-1; encoded by the PDCD1 gene) receptor expressed on activated T cells and its ligand, programmed death-ligand 1 (PD-L1; encoded by the CD274 gene), is a major co-inhibitory checkpoint signaling that controls T cell activities. Various types of cancers express high levels of PD-L1 and exploit PD-L1/PD-1 signaling to evade T cell immunity. Blocking the PD-L1/PD-1 pathway has consistently shown remarkable anti-tumor effects in patients with advanced cancers and is recognized as the gold standard for developing new immune checkpoint blockade (ICB) and combination therapies. However, the response rates of anti-PD-L1 have been limited in several solid tumors. Therefore, furthering our understanding of the regulatory mechanisms of PD-L1 can bring substantial benefits to patients with cancer by improving the efficacy of current PD-L1/PD-1 blockade or other ICBs. In this review, we provide current knowledge of PD-L1 regulatory mechanisms at the transcriptional, posttranscriptional, post-translational, and extracellular levels, and discuss the implications of these findings in cancer diagnosis and immunotherapy.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Protein Processing, Post-Translational , RNA Processing, Post-Transcriptional , Signal Transduction , Transcription, Genetic , Tumor Escape
12.
Semin Immunol ; 70: 101833, 2023 11.
Article in English | MEDLINE | ID: mdl-37647772

ABSTRACT

The identification of gasdermin as the executor of pyroptosis has opened new avenues for the study of this process. Although pyroptosis research has mainly focused on immune cells since it was discovered three decades ago, accumulating evidence suggests that pyroptosis plays crucial roles in many biological processes. One example is the discovery of gasdermin-mediated cancer cell pyroptosis (CCP) which has become an important and frontier field in oncology. Recent studies have shown that CCP induction can heat tumor microenvironment (TME) and thereby elicit the robust anti-tumor immunity to suppress tumor growth. As a newly discovered form of tumor cell death, CCP offers promising opportunities for improving tumor treatment and developing new drugs. Nevertheless, the research on CCP is still in its infancy, and the molecular mechanisms underlying the expression, regulation and activation of gasdermins are not yet fully understood. In this review, we summarize the recent progress of gasdermin research in cancer area, and propose that the anti-tumor effect of immune cell pyroptosis (ICP) and CCP depends on their duration, intensity, and the type of cells undergoing pyroptosis within TME.


Subject(s)
Gasdermins , Neoplasms , Humans , Neoplasms/therapy , Carcinogenesis , Tumor Microenvironment , Pyroptosis
13.
Proc Natl Acad Sci U S A ; 121(28): e2322972121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968116

ABSTRACT

Rapid accumulation of repair factors at DNA double-strand breaks (DSBs) is essential for DSB repair. Several factors involved in DSB repair have been found undergoing liquid-liquid phase separation (LLPS) at DSB sites to facilitate DNA repair. RNF168, a RING-type E3 ubiquitin ligase, catalyzes H2A.X ubiquitination for recruiting DNA repair factors. Yet, whether RNF168 undergoes LLPS at DSB sites remains unclear. Here, we identified K63-linked polyubiquitin-triggered RNF168 condensation which further promoted RNF168-mediated DSB repair. RNF168 formed liquid-like condensates upon irradiation in the nucleus while purified RNF168 protein also condensed in vitro. An intrinsically disordered region containing amino acids 460-550 was identified as the essential domain for RNF168 condensation. Interestingly, LLPS of RNF168 was significantly enhanced by K63-linked polyubiquitin chains, and LLPS largely enhanced the RNF168-mediated H2A.X ubiquitination, suggesting a positive feedback loop to facilitate RNF168 rapid accumulation and its catalytic activity. Functionally, LLPS deficiency of RNF168 resulted in delayed recruitment of 53BP1 and BRCA1 and subsequent impairment in DSB repair. Taken together, our finding demonstrates the pivotal effect of LLPS in RNF168-mediated DSB repair.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Humans , Tumor Suppressor p53-Binding Protein 1/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Ubiquitin/metabolism , Histones/metabolism , Histones/genetics , Polyubiquitin/metabolism
14.
Nature ; 580(7804): 530-535, 2020 04.
Article in English | MEDLINE | ID: mdl-32322062

ABSTRACT

Cancer cells increase lipogenesis for their proliferation and the activation of sterol regulatory element-binding proteins (SREBPs) has a central role in this process. SREBPs are inhibited by a complex composed of INSIG proteins, SREBP cleavage-activating protein (SCAP) and sterols in the endoplasmic reticulum. Regulation of the interaction between INSIG proteins and SCAP by sterol levels is critical for the dissociation of the SCAP-SREBP complex from the endoplasmic reticulum and the activation of SREBPs1,2. However, whether this protein interaction is regulated by a mechanism other than the abundance of sterol-and in particular, whether oncogenic signalling has a role-is unclear. Here we show that activated AKT in human hepatocellular carcinoma (HCC) cells phosphorylates cytosolic phosphoenolpyruvate carboxykinase 1 (PCK1), the rate-limiting enzyme in gluconeogenesis, at Ser90. Phosphorylated PCK1 translocates to the endoplasmic reticulum, where it uses GTP as a phosphate donor to phosphorylate INSIG1 at Ser207 and INSIG2 at Ser151. This phosphorylation reduces the binding of sterols to INSIG1 and INSIG2 and disrupts the interaction between INSIG proteins and SCAP, leading to the translocation of the SCAP-SREBP complex to the Golgi apparatus, the activation of SREBP proteins (SREBP1 or SREBP2) and the transcription of downstream lipogenesis-related genes, proliferation of tumour cells, and tumorigenesis in mice. In addition, phosphorylation of PCK1 at Ser90, INSIG1 at Ser207 and INSIG2 at Ser151 is not only positively correlated with the nuclear accumulation of SREBP1 in samples from patients with HCC, but also associated with poor HCC prognosis. Our findings highlight the importance of the protein kinase activity of PCK1 in the activation of SREBPs, lipogenesis and the development of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gluconeogenesis , Intracellular Signaling Peptides and Proteins/metabolism , Lipogenesis , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Animals , Carcinogenesis , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Liver Neoplasms/pathology , Male , Membrane Proteins/chemistry , Mice , Mice, Nude , Oxysterols/metabolism , Phosphorylation , Prognosis , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism
15.
Mol Cell ; 71(4): 606-620.e7, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30118680

ABSTRACT

Metformin has been reported to possess antitumor activity and maintain high cytotoxic T lymphocyte (CTL) immune surveillance. However, the functions and detailed mechanisms of metformin's role in cancer immunity are not fully understood. Here, we show that metformin increases CTL activity by reducing the stability and membrane localization of programmed death ligand-1 (PD-L1). Furthermore, we discover that AMP-activated protein kinase (AMPK) activated by metformin directly phosphorylates S195 of PD-L1. S195 phosphorylation induces abnormal PD-L1 glycosylation, resulting in its ER accumulation and ER-associated protein degradation (ERAD). Consistently, tumor tissues from metformin-treated breast cancer patients exhibit reduced PD-L1 levels with AMPK activation. Blocking the inhibitory signal of PD-L1 by metformin enhances CTL activity against cancer cells. Our findings identify a new regulatory mechanism of PD-L1 expression through the ERAD pathway and suggest that the metformin-CTLA4 blockade combination has the potential to increase the efficacy of immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , B7-H1 Antigen/genetics , CTLA-4 Antigen/genetics , Gene Expression Regulation, Neoplastic , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Animals , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Female , Glycosylation , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Mammary Glands, Human/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred NOD , Phosphorylation , Serine/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
16.
Mol Cell ; 69(2): 279-291.e5, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29351847

ABSTRACT

Sustained energy starvation leads to activation of AMP-activated protein kinase (AMPK), which coordinates energy status with numerous cellular processes including metabolism, protein synthesis, and autophagy. Here, we report that AMPK phosphorylates the histone methyltransferase EZH2 at T311 to disrupt the interaction between EZH2 and SUZ12, another core component of the polycomb repressive complex 2 (PRC2), leading to attenuated PRC2-dependent methylation of histone H3 at Lys27. As such, PRC2 target genes, many of which are known tumor suppressors, were upregulated upon T311-EZH2 phosphorylation, which suppressed tumor cell growth both in cell culture and mouse xenografts. Pathologically, immunohistochemical analyses uncovered a positive correlation between AMPK activity and pT311-EZH2, and higher pT311-EZH2 correlates with better survival in both ovarian and breast cancer patients. Our finding suggests that AMPK agonists might be promising sensitizers for EZH2-targeting cancer therapies.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Animals , Carcinogenesis/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , DNA Methylation , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/physiology , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Mice , Neoplasm Proteins , Nuclear Proteins/metabolism , Oncogenes , Ovarian Neoplasms/metabolism , Phosphorylation , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/physiology , Transcription Factors , Up-Regulation
17.
Nucleic Acids Res ; 52(1): 154-165, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-37986225

ABSTRACT

Asymmetric cell division (ACD) is a mechanism used by stem cells to maintain the number of progeny. However, the epigenetic mechanisms regulating ACD remain elusive. Here we show that BRD4, a BET domain protein that binds to acetylated histone, is segregated in daughter cells together with H3K56Ac and regulates ACD. ITGB1 is regulated by BRD4 to regulate ACD. A long noncoding RNA (lncRNA), LIBR (LncRNA Inhibiting BRD4), decreases the percentage of stem cells going through ACD through interacting with the BRD4 mRNAs. LIBR inhibits the translation of BRD4 through recruiting a translation repressor, RCK, and inhibiting the binding of BRD4 mRNAs to polysomes. These results identify the epigenetic regulatory modules (BRD4, lncRNA LIBR) that regulate ACD. The regulation of ACD by BRD4 suggests the therapeutic limitation of using BRD4 inhibitors to treat cancer due to the ability of these inhibitors to promote symmetric cell division that may lead to tumor progression and treatment resistance.


Subject(s)
Bromodomain Containing Proteins , Cell Division , Epigenesis, Genetic , RNA, Long Noncoding , Asymmetric Cell Division , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Bromodomain Containing Proteins/metabolism
18.
Nucleic Acids Res ; 52(D1): D1246-D1252, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37956338

ABSTRACT

Advancements in high-throughput technology offer researchers an extensive range of multi-omics data that provide deep insights into the complex landscape of cancer biology. However, traditional statistical models and databases are inadequate to interpret these high-dimensional data within a multi-omics framework. To address this limitation, we introduce DriverDBv4, an updated iteration of the DriverDB cancer driver gene database (http://driverdb.bioinfomics.org/). This updated version offers several significant enhancements: (i) an increase in the number of cohorts from 33 to 70, encompassing approximately 24 000 samples; (ii) inclusion of proteomics data, augmenting the existing types of omics data and thus expanding the analytical scope; (iii) implementation of multiple multi-omics algorithms for identification of cancer drivers; (iv) new visualization features designed to succinctly summarize high-context data and redesigned existing sections to accommodate the increased volume of datasets and (v) two new functions in Customized Analysis, specifically designed for multi-omics driver identification and subgroup expression analysis. DriverDBv4 facilitates comprehensive interpretation of multi-omics data across diverse cancer types, thereby enriching the understanding of cancer heterogeneity and aiding in the development of personalized clinical approaches. The database is designed to foster a more nuanced understanding of the multi-faceted nature of cancer.


Subject(s)
Databases, Genetic , Multiomics , Neoplasms , Humans , Algorithms , Databases, Genetic/standards , Neoplasms/genetics , Neoplasms/physiopathology
19.
Nucleic Acids Res ; 52(W1): W390-W397, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38709887

ABSTRACT

In the field of lipidomics, where the complexity of lipid structures and functions presents significant analytical challenges, LipidSig stands out as the first web-based platform providing integrated, comprehensive analysis for efficient data mining of lipidomic datasets. The upgraded LipidSig 2.0 (https://lipidsig.bioinfomics.org/) simplifies the process and empowers researchers to decipher the complex nature of lipids and link lipidomic data to specific characteristics and biological contexts. This tool markedly enhances the efficiency and depth of lipidomic research by autonomously identifying lipid species and assigning 29 comprehensive characteristics upon data entry. LipidSig 2.0 accommodates 24 data processing methods, streamlining diverse lipidomic datasets. The tool's expertise in automating intricate analytical processes, including data preprocessing, lipid ID annotation, differential expression, enrichment analysis, and network analysis, allows researchers to profoundly investigate lipid properties and their biological implications. Additional innovative features, such as the 'Network' function, offer a system biology perspective on lipid interactions, and the 'Multiple Group' analysis aids in examining complex experimental designs. With its comprehensive suite of features for analyzing and visualizing lipid properties, LipidSig 2.0 positions itself as an indispensable tool for advanced lipidomics research, paving the way for new insights into the role of lipids in cellular processes and disease development.


Subject(s)
Lipidomics , Lipids , Software , Lipids/chemistry , Lipidomics/instrumentation , Lipidomics/methods , Data Analysis , Internet , Algorithms , Data Visualization
20.
Trends Genet ; 38(10): 991-995, 2022 10.
Article in English | MEDLINE | ID: mdl-35637074

ABSTRACT

Polyploidization and polyploidy reversal (depolyploidization) are crucial pathways to conversely alter genomic contents in organisms. Understanding the mechanisms switching between polyploidization and polyploidy reversal should broaden our knowledge of the generation of pathological polyploidy and pave a new path to prevent related diseases.


Subject(s)
Mitosis , Polyploidy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL