Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Proc Natl Acad Sci U S A ; 120(50): e2315163120, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38055744

ABSTRACT

Interferon-induced ubiquitin (Ub)-like modifier ISG15 covalently modifies host and viral proteins to restrict viral infections. Its function is counteracted by the canonical deISGylase USP18 or Ub-specific protease 18. Notwithstanding indications for the existence of other ISG15 cross-reactive proteases, these remain to be identified. Here, we identify deubiquitinase USP16 as an ISG15 cross-reactive protease by means of ISG15 activity-based profiling. Recombinant USP16 cleaved pro-ISG15 and ISG15 isopeptide-linked model substrates in vitro, as well as ISGylated substrates from cell lysates. Moreover, interferon-induced stimulation of ISGylation was increased by depletion of USP16. The USP16-dependent ISG15 interactome indicated that the deISGylating function of USP16 may regulate metabolic pathways. Targeted enzymes include malate dehydrogenase, cytoplasmic superoxide dismutase 1, fructose-bisphosphate aldolase A, and cytoplasmic glutamic-oxaloacetic transaminase 1. USP16 may thus contribute to the regulation of a subset of metabolism-related proteins during type-I interferon responses.


Subject(s)
Cytokines , Interferon Type I , Cytokines/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Peptide Hydrolases/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Deubiquitinating Enzymes
2.
ACS Chem Biol ; 18(9): 2003-2013, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37642399

ABSTRACT

Ubiquitin thioesterase OTUB2, a cysteine protease from the ovarian tumor (OTU) deubiquitinase superfamily, is often overexpressed during tumor progression and metastasis. Development of OTUB2 inhibitors is therefore believed to be therapeutically important, yet potent and selective small-molecule inhibitors targeting OTUB2 are scarce. Here, we describe the development of an improved OTUB2 inhibitor, LN5P45, comprising a chloroacethydrazide moiety that covalently reacts to the active-site cysteine residue. LN5P45 shows outstanding target engagement and proteome-wide selectivity in living cells. Importantly, LN5P45 as well as other OTUB2 inhibitors strongly induce monoubiquitination of OTUB2 on lysine 31. We present a route to future OTUB2-related therapeutics and have shown that the OTUB2 inhibitor developed in this study can help to uncover new aspects of the related biology and open new questions regarding the understanding of OTUB2 regulation at the post-translational modification level.


Subject(s)
Cysteine Proteases , Protein Processing, Post-Translational , Ubiquitination , Ubiquitin , Cysteine
3.
Angew Chem Int Ed Engl ; 62(32): e202303319, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37272691

ABSTRACT

Protein post-translational modification with ubiquitin (Ub) is a versatile signal regulating almost all aspects of cell biology, and an increasing range of diseases is associated with impaired Ub modification. In this light, the Ub system offers an attractive, yet underexplored route to the development of novel targeted treatments. A promising strategy for small molecule intervention is posed by the final components of the enzymatic ubiquitination cascade, E3 ligases, as they determine the specificity of the protein ubiquitination pathway. Here, we present UbSRhodol, an autoimmolative Ub-based probe, which upon E3 processing liberates the pro-fluorescent dye, amenable to profile the E3 transthiolation activity for recombinant and in cell-extract E3 ligases. UbSRhodol enabled detection of changes in transthiolation efficacy evoked by enzyme key point mutations or conformational changes, and offers an excellent assay reagent amenable to a high-throughput screening setup allowing the identification of small molecules modulating E3 activity.


Subject(s)
Fluorescent Dyes , Ubiquitin , Ubiquitin/metabolism , Cysteine/metabolism , Ubiquitination , Ubiquitin-Protein Ligases/metabolism
4.
Nat Commun ; 13(1): 4461, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35915084

ABSTRACT

Cancer immunotherapies have shown clinical success in various types of tumors but the patient response rate is low, particularly in breast cancer. Here we report that malignant breast cancer cells can transfer active TGF-ß type II receptor (TßRII) via tumor-derived extracellular vesicles (TEV) and thereby stimulate TGF-ß signaling in recipient cells. Up-take of extracellular vesicle-TßRII (EV-TßRII) in low-grade tumor cells initiates epithelial-to-mesenchymal transition (EMT), thus reinforcing cancer stemness and increasing metastasis in intracardial xenograft and orthotopic transplantation models. EV-TßRII delivered as cargo to CD8+ T cells induces the activation of SMAD3 which we demonstrated to associate and cooperate with TCF1 transcription factor to impose CD8+ T cell exhaustion, resulting in failure of immunotherapy. The levels of TßRII+ circulating extracellular vesicles (crEV) appears to correlate with tumor burden, metastasis and patient survival, thereby serve as a non-invasive screening tool to detect malignant breast tumor stages. Thus, our findings not only identify a possible mechanism by which breast cancer cells can promote T cell exhaustion and dampen host anti-tumor immunity, but may also identify a target for immune therapy against the most devastating breast tumors.


Subject(s)
Breast Neoplasms , Extracellular Vesicles , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Extracellular Vesicles/metabolism , Female , Humans , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
5.
Oncogene ; 39(22): 4436-4449, 2020 05.
Article in English | MEDLINE | ID: mdl-32350443

ABSTRACT

Activator protein (AP)-1 transcription factors are essential elements of the pro-oncogenic functions of transforming growth factor-ß (TGFß)-SMAD signaling. Here we show that in multiple HER2+ and/or EGFR+ breast cancer cell lines these AP-1-dependent tumorigenic properties of TGFß critically rely on epidermal growth factor receptor (EGFR) activation and expression of the ΔN isoform of transcriptional regulator p63. EGFR and ΔNp63 enabled and/or potentiated the activation of a subset of TGFß-inducible invasion/migration-associated genes, e.g., ITGA2, LAMB3, and WNT7A/B, and enhanced the recruitment of SMAD2/3 to these genes. The TGFß- and EGF-induced binding of SMAD2/3 and JUNB to these gene loci was accompanied by p63-SMAD2/3 and p63-JUNB complex formation. p63 and EGFR were also found to strongly potentiate TGFß induction of AP-1 proteins and, in particular, FOS family members. Ectopic overexpression of FOS could counteract the decrease in TGFß-induced gene activation after p63 depletion. p63 is also involved in the transcriptional regulation of heparin binding (HB)-EGF and EGFR genes, thereby establishing a self-amplification loop that facilitates and empowers the pro-invasive functions of TGFß. These cooperative pro-oncogenic functions of EGFR, AP-1, p63, and TGFß were efficiently inhibited by clinically relevant chemical inhibitors. Our findings may, therefore, be of importance for therapy of patients with breast cancers with an activated EGFR-RAS-RAF pathway.


Subject(s)
Breast Neoplasms/pathology , Epidermal Growth Factor/physiology , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Neoplasm Proteins/physiology , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Transcription, Genetic , Transforming Growth Factor beta1/physiology , Tumor Suppressor Proteins/genetics , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , ErbB Receptors/physiology , Female , Humans , MAP Kinase Signaling System , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-fos/physiology , Proto-Oncogene Proteins c-jun/physiology , Receptor, ErbB-2/physiology , Receptor, Transforming Growth Factor-beta Type I/physiology , Smad Proteins/physiology
6.
J Mol Cell Biol ; 12(2): 138-151, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31291647

ABSTRACT

The transforming growth factor-ß (TGF-ß) family controls embryogenesis, stem cell differentiation, and tissue homeostasis. However, how post-translation modifications contribute to fine-tuning of TGF-ß family signaling responses is not well understood. Inhibitory (I)-Smads can antagonize TGF-ß/Smad signaling by recruiting Smurf E3 ubiquitin ligases to target the active TGF-ß receptor for proteasomal degradation. A proteomic interaction screen identified Vpr binding protein (VprBP) as novel binding partner of Smad7. Mis-expression studies revealed that VprBP negatively controls Smad2 phosphorylation, Smad2-Smad4 interaction, as well as TGF-ß target gene expression. VprBP was found to promote Smad7-Smurf1-TßRI complex formation and induce proteasomal degradation of TGF-ß type I receptor (TßRI). Moreover, VprBP appears to stabilize Smurf1 by suppressing Smurf1 poly-ubiquitination. In multiple adult and mouse embryonic stem cells, depletion of VprBP promotes TGF-ß or Activin-induced responses. In the mouse embryo VprBP expression negatively correlates with mesoderm marker expression, and VprBP attenuated mesoderm induction during zebrafish embryogenesis. Our findings thereby uncover a novel regulatory mechanism by which Smurf1 controls the TGF-ß and Activin cascade and identify VprBP as a critical determinant of embryonic mesoderm induction.


Subject(s)
Activins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Receptor, Transforming Growth Factor-beta Type I/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Activins/pharmacology , Animals , Embryonic Development/drug effects , Embryonic Development/genetics , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , HEK293 Cells , HaCaT Cells , Hep G2 Cells , Humans , Mesoderm/embryology , Mesoderm/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transfection , Transforming Growth Factor beta/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Zebrafish/embryology
7.
Cells ; 8(12)2019 11 21.
Article in English | MEDLINE | ID: mdl-31766464

ABSTRACT

Transforming growth factor-ß (TGFß) has both tumor-suppressive and tumor-promoting effects in breast cancer. These functions are partly mediated through Smads, intracellular transcriptional effectors of TGFß. Smads form complexes with other DNA-binding transcription factors to elicit cell-type-dependent responses. Previously, we found that the collagen invasion and migration of pre-malignant breast cancer cells in response to TGFß and epidermal growth factor (EGF) critically depend on multiple Jun and Fos components of the activator protein (AP)-1 transcription factor complex. Here we report that the same process is negatively regulated by Jun N-terminal kinase (JNK)-dependent cJun phosphorylation. This was demonstrated by analysis of phospho-deficient, phospho-mimicking, and dimer-specific cJun mutants, and experiments employing a mutant version of the phosphatase MKP1 that specifically inhibits JNK. Hyper-phosphorylation of cJun by JNK strongly inhibited its ability to induce several Jun/Fos-regulated genes and to promote migration and invasion. These results show that MEK-AP-1 and JNK-phospho-cJun exhibit distinct pro- and anti-invasive functions, respectively, through differential regulation of Smad- and AP-1-dependent TGFß target genes. Our findings are of importance for personalized cancer therapy, such as for patients suffering from specific types of breast tumors with activated EGF receptor-Ras or inactivated JNK pathways.


Subject(s)
Breast Neoplasms/metabolism , Epidermal Growth Factor/pharmacology , MAP Kinase Signaling System , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genes, jun , HeLa Cells , Humans , Neoplasm Invasiveness , Phosphorylation , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Transcription Factor AP-1/genetics
8.
Breast Cancer Res ; 21(1): 109, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31533776

ABSTRACT

BACKGROUND: Bone morphogenetic proteins (BMPs) have been reported to maintain epithelial integrity and to antagonize the transforming growth factor ß (TGFß)-induced epithelial to mesenchymal transition. The expression of soluble BMP antagonists is dysregulated in cancers and interrupts proper BMP signaling in breast cancer. METHODS: In this study, we mined the prognostic role of BMP antagonists GREMLIN 1 (GREM1) in primary breast cancer tissues using in-house and publicly available datasets. We determined which cells express GREM1 RNA using in situ hybridization (ISH) on a breast cancer tissue microarray. The effects of Grem1 on the properties of breast cancer cells were assessed by measuring the mesenchymal/stem cell marker expression and functional cell-based assays for stemness and invasion. The role of Grem1 in breast cancer-associated fibroblast (CAF) activation was measured by analyzing the expression of fibroblast markers, phalloidin staining, and collagen contraction assays. The role of Grem1 in CAF-induced breast cancer cell intravasation and extravasation was studied by utilizing xenograft zebrafish breast cancer (co-) injection models. RESULTS: Expression analysis of clinical breast cancer datasets revealed that high expression of GREM1 in breast cancer stroma is correlated with a poor prognosis regardless of the molecular subtype. The large majority of human breast cancer cell lines did not express GREM1 in vitro, but breast CAFs did express GREM1 both in vitro and in vivo. Transforming growth factor ß (TGFß) secreted by breast cancer cells, and also inflammatory cytokines, stimulated GREM1 expression in CAFs. Grem1 abrogated bone morphogenetic protein (BMP)/SMAD signaling in breast cancer cells and promoted their mesenchymal phenotype, stemness, and invasion. Moreover, Grem1 production by CAFs strongly promoted the fibrogenic activation of CAFs and promoted breast cancer cell intravasation and extravasation in co-injection xenograft zebrafish models. CONCLUSIONS: Our results demonstrated that Grem1 is a pivotal factor in the reciprocal interplay between breast cancer cells and CAFs, which promotes cancer cell invasion. Targeting Grem1 could be beneficial in the treatment of breast cancer patients with high Grem1 expression.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Cytokines/metabolism , Disease Progression , Epithelial-Mesenchymal Transition , Female , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mammary Neoplasms, Experimental , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Phosphorylation , Prognosis , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Zebrafish
9.
J Pathol ; 247(3): 333-346, 2019 03.
Article in English | MEDLINE | ID: mdl-30430573

ABSTRACT

Endothelial-to-mesenchymal transition (EndMT) has been unveiled as a common cause for a multitude of human pathologies, including cancer and cardiovascular disease. Vascular calcification is a risk factor for ischemic vascular disorders and slowing calcification may reduce mortality in affected patients. The absence of early biomarkers hampers the identification of patients at risk. EndMT and vascular calcification are induced upon cooperation between distinct stimuli, including inflammatory cytokines and transforming growth factor beta (TGF-ß) family members. However, how these signaling pathways interplay to promote cell differentiation and eventually vascular calcification is not well understood. Using in vitro and ex vivo analysis in animal models and patient-derived tissues, we have identified that the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1ß) induce EndMT in human primary aortic endothelial cells, thereby sensitizing them for BMP-9-induced osteogenic differentiation. Downregulation of the BMP type II receptor BMPR2 is a key event in this process. Rather than compromising BMP canonical signal transduction, loss of BMPR2 results in decreased JNK signaling in ECs, thus enhancing BMP-9-induced mineralization. Altogether, our results point at the BMPR2-JNK signaling axis as a key pathway regulating inflammation-induced EndMT and contributing to calcification. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/physiology , Epithelial-Mesenchymal Transition/physiology , Vascular Calcification/physiopathology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/physiology , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Epithelial-Mesenchymal Transition/drug effects , Humans , Inflammation Mediators/pharmacology , Interleukin-1beta/pharmacology , Mice, Inbred C3H , Osteogenesis/drug effects , Osteogenesis/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Calcification/pathology
10.
Mol Cell ; 73(1): 7-21.e7, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30472188

ABSTRACT

The transcriptional regulators YAP and TAZ play important roles in development, physiology, and tumorigenesis and are negatively controlled by the Hippo pathway. It is yet unknown why the YAP/ TAZ proteins are frequently activated in human malignancies in which the Hippo pathway is still active. Here, by a gain-of-function cancer metastasis screen, we discovered OTUB2 as a cancer stemness and metastasis-promoting factor that deubiquitinates and activates YAP/TAZ. We found OTUB2 to be poly-SUMOylated on lysine 233, and this SUMOylation enables it to bind YAP/TAZ. We also identified a yet-unknown SUMO-interacting motif (SIM) in YAP and TAZ required for their association with SUMOylated OTUB2. Importantly, EGF and oncogenic KRAS induce OTUB2 poly-SUMOylation and thereby activate YAP/TAZ. Our results establish OTUB2 as an essential modulator of YAP/TAZ and also reveal a novel mechanism via which YAP/TAZ activity is induced by oncogenic KRAS.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/enzymology , Cell Movement , Intracellular Signaling Peptides and Proteins/metabolism , Neoplastic Stem Cells/enzymology , Phosphoproteins/metabolism , Thiolester Hydrolases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Movement/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/agonists , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lysine , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Phenotype , Phosphoproteins/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Sumoylation , Thiolester Hydrolases/genetics , Time Factors , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
11.
Cell Host Microbe ; 24(6): 776-790.e5, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30472208

ABSTRACT

Mitochondrial antiviral signaling protein (MAVS) is an adaptor of the innate immune receptor retinoic acid-inducible gene 1 (RIG-I) that links recognition of viral RNA to antiviral signaling. Upon interacting with RIG-I, MAVS undergoes lysine 63-linked poly-ubiquitination by the E3 ligase TRIM31 and subsequently aggregates to activate downstream signaling effectors. We find that the scaffold protein FAF1 forms aggregates that negatively regulate MAVS. FAF1 antagonizes the poly-ubiquitination and aggregation of MAVS by competing with TRIM31 for MAVS association. FAF1 knockout mice are more resistant to RNA virus infection, and FAF1 deficiency in myeloid cells results in enhanced innate signaling and reduced viral load and morbidity in vivo. Upon virus infection, the kinase IKKɛ directly phosphorylates FAF1 at Ser556 and triggers FAF1 de-aggregation. Moreover, Ser556 phosphorylation promotes FAF1 lysosomal degradation, consequently relieving FAF1-dependent suppression of MAVS. These findings establish FAF1 as a modulator of MAVS and uncover mechanisms that regulate FAF1 to insure timely activation of antiviral defense.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunity, Innate/immunology , Phosphorylation/immunology , RNA Virus Infections/immunology , A549 Cells , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Mice, Knockout , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
12.
Oncogene ; 37(48): 6195-6211, 2018 11.
Article in English | MEDLINE | ID: mdl-30002444

ABSTRACT

Chemoresistance remains a major complication of cancer treatments. Recent data provide strong evidence that chemoresistance is linked to epithelial-mesenchymal transition (EMT), a latent developmental process, which is re-activated during cancer progression. EMT involves transcriptional reprogramming and is driven by specific EMT transcription factors (EMT-TFs). In this review, we provide support for the idea that EMT-TFs contribute to the development of resistance against cancer therapy and discuss how EMT-TFs might be targeted to advance novel therapeutic approaches to the treatment of cancer.


Subject(s)
Drug Resistance, Neoplasm/physiology , Epithelial-Mesenchymal Transition/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Transcription Factors/metabolism , Animals , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/physiology , Humans
13.
Nat Immunol ; 19(3): 233-245, 2018 03.
Article in English | MEDLINE | ID: mdl-29358709

ABSTRACT

Malignancies can compromise innate immunity, but the mechanisms of this are largely unknown. Here we found that, via tumor-derived exosomes (TEXs), cancers were able to transfer activated epidermal growth factor receptor (EGFR) to host macrophages and thereby suppress innate antiviral immunity. Screening of the human kinome identified the kinase MEKK2 in macrophages as an effector of TEX-delivered EGFR that negatively regulated the antiviral immune response. In the context of experimental tumor implantation, MEKK2-deficient mice were more resistant to viral infection than were wild-type mice. Injection of TEXs into mice reduced innate immunity, increased viral load and increased morbidity in an EGFR- and MEKK2-dependent manner. MEKK2 phosphorylated IRF3, a transcription factor crucial for the production of type I interferons; this triggered poly-ubiquitination of IRF3 and blocked its dimerization, translocation to the nucleus and transcriptional activity after viral infection. These findings identify a mechanism by which cancer cells can dampen host innate immunity and potentially cause patients with cancer to become immunocompromised.


Subject(s)
ErbB Receptors/immunology , Exosomes/immunology , Immunity, Innate/immunology , Neoplasms/immunology , Virus Diseases/immunology , Adult , Animals , ErbB Receptors/metabolism , Exosomes/metabolism , Female , Humans , Immunocompromised Host/immunology , MAP Kinase Kinase Kinase 2/immunology , MAP Kinase Kinase Kinase 2/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged
14.
Nucleic Acids Res ; 46(3): 1180-1195, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29186616

ABSTRACT

It is well established that transforming growth factor-ß (TGFß) switches its function from being a tumor suppressor to a tumor promoter during the course of tumorigenesis, which involves both cell-intrinsic and environment-mediated mechanisms. We are interested in breast cancer cells, in which SMAD mutations are rare and interactions between SMAD and other transcription factors define pro-oncogenic events. Here, we have performed chromatin immunoprecipitation (ChIP)-sequencing analyses which indicate that the genome-wide landscape of SMAD2/3 binding is altered after prolonged TGFß stimulation. De novo motif analyses of the SMAD2/3 binding regions predict enrichment of binding motifs for activator protein (AP)1 in addition to SMAD motifs. TGFß-induced expression of the AP1 component JUNB was required for expression of many late invasion-mediating genes, creating a feed-forward regulatory network. Moreover, we found that several components in the WNT pathway were enriched among the late TGFß-target genes, including the invasion-inducing WNT7 proteins. Consistently, overexpression of WNT7A or WNT7B enhanced and potentiated TGFß-induced breast cancer cell invasion, while inhibition of the WNT pathway reduced this process. Our study thereby helps to explain how accumulation of pro-oncogenic stimuli switches and stabilizes TGFß-induced cellular phenotypes of epithelial cells.


Subject(s)
Breast Neoplasms/genetics , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Transcription Factors/genetics , Transforming Growth Factor beta1/pharmacology , Animals , Base Sequence , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Embryo, Nonmammalian , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Neoplasm Invasiveness , Protein Binding , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zebrafish
15.
Acta Biochim Biophys Sin (Shanghai) ; 50(1): 121-132, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29190313

ABSTRACT

The transforming growth factor (TGF)-ß signaling events are well known to control diverse processes and numerous responses, such as cell proliferation, differentiation, apoptosis, and migration. TGF-ß signaling plays context-dependent roles in cancer: in pre-malignant cells TGF-ß primarily functions as a tumor suppressor, while in the later stages of cancer TGF-ß signaling promotes invasion and metastasis. Recent studies have also suggested that the cross-talk between TGF-ß signaling and other signaling pathways, such as Hippo, Wnt, EGFR/RAS, and PI3K/AKT pathways, may substantially contribute to our current understanding of TGF-ß signaling and cancer. As a result of the wide-ranging effects of TGF-ß, blockade of TGF-ß and its downstream signaling components provides multiple therapeutic opportunities. Therefore, the outlook for anti-TGF-ß signaling therapy for numerous diseases appears bright and will provide valuable information and thinking on the drug molecular design. In this review, we focus on recent insights into the regulation of TGF-ß signaling in cancer metastasis which may contribute to the development of novel cancer-targeting therapies.


Subject(s)
Neoplasms/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/therapeutic use , Disease Progression , Humans , Models, Biological , Molecular Targeted Therapy/methods , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors
16.
Nat Commun ; 8(1): 2116, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235476

ABSTRACT

Metastasis is the main cause of death in cancer patients. TGF-ß is pro-metastatic for malignant cancer cells. Here we report a loss-of-function screen in mice with metastasis as readout and identify OTUD1 as a metastasis-repressing factor. OTUD1-silenced cancer cells show mesenchymal and stem-cell-like characteristics. Further investigation reveals that OTUD1 directly deubiquitinates the TGF-ß pathway inhibitor SMAD7 and prevents its degradation. Moreover, OTUD1 cleaves Lysine 33-linked poly-ubiquitin chains of SMAD7 Lysine 220, which exposes the SMAD7 PY motif, enabling SMURF2 binding and subsequent TßRI turnover at the cell surface. Importantly, OTUD1 is lost in multiple types of human cancers and loss of OTUD1 increases metastasis in intracardial xenograft and orthotopic transplantation models, and correlates with poor prognosis among breast cancer patients. High levels of OTUD1 inhibit cancer stemness and shut off metastasis. Thus, OTUD1 represses breast cancer metastasis by mitigating TGF-ß-induced pro-oncogenic responses via deubiquitination of SMAD7.


Subject(s)
Breast Neoplasms/metabolism , Smad7 Protein/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line , Cell Line, Tumor , Doxycycline/pharmacology , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , RNA Interference , Smad7 Protein/genetics , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays
18.
Nat Immunol ; 18(7): 733-743, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28481329

ABSTRACT

The transcription regulator YAP controls organ size by regulating cell growth, proliferation and apoptosis. However, whether YAP has a role in innate antiviral immunity is largely unknown. Here we found that YAP negatively regulated an antiviral immune response. YAP deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in vivo. YAP blocked dimerization of the transcription factor IRF3 and impeded translocation of IRF3 to the nucleus after viral infection. Notably, virus-activated kinase IKKɛ phosphorylated YAP at Ser403 and thereby triggered degradation of YAP in lysosomes and, consequently, relief of YAP-mediated inhibition of the cellular antiviral response. These findings not only establish YAP as a modulator of the activation of IRF3 but also identify a previously unknown regulatory mechanism independent of the kinases Hippo and LATS via which YAP is controlled by the innate immune pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Fibroblasts/immunology , I-kappa B Kinase/metabolism , Immunity, Innate/immunology , Lysosomes/metabolism , Macrophages/immunology , Phosphoproteins/immunology , Rhabdoviridae Infections/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CRISPR-Cas Systems , Cell Cycle Proteins , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Fluorescent Antibody Technique , Gene Editing , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Lung/immunology , Lung/pathology , Mice , Microscopy, Confocal , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdoviridae Infections/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Vesiculovirus , Viral Load , YAP-Signaling Proteins
19.
Nat Commun ; 8: 15021, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28443643

ABSTRACT

TGF-ß is pro-metastatic for the late-stage breast cancer cells. Despite recent progress, the regulation of TGF-ß type II receptor remains uncertain. Here we report that FAF1 destabilizes TßRII on the cell surface by recruiting the VCP/E3 ligase complex, thereby limiting excessive TGF-ß response. Importantly, activated AKT directly phosphorylates FAF1 at Ser 582, which disrupts the FAF1-VCP complex and reduces FAF1 at the plasma membrane. The latter results in an increase in TßRII at the cell surface that promotes both TGF-ß-induced SMAD and non-SMAD signalling. We uncover a metastasis suppressing role for FAF1 through analyses of FAF1-knockout animals, various in vitro and in vivo models of epithelial-to-mesenchymal transition and metastasis, an MMTV-PyMT transgenic mouse model of mammary tumour progression and clinical breast cancer samples. These findings describe a previously uncharacterized mechanism by which TßRII is tightly controlled. Together, we reveal how SMAD and AKT pathways interact to confer pro-oncogenic responses to TGF-ß.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Transforming Growth Factor beta/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Metastasis , Phosphorylation , Receptor, Transforming Growth Factor-beta Type II , Transplantation, Heterologous
20.
J Pathol ; 241(5): 567-577, 2017 04.
Article in English | MEDLINE | ID: mdl-28035672

ABSTRACT

Metastasis of malignant cells to vital organs remains the major cause of mortality in many types of cancers. The tumour invasion-metastasis cascade is a stepwise and multistage process whereby tumour cells disseminate from primary sites and spread to colonize distant sites through the systemic haematogenous or lymphatic circulations. The general steps of metastasis may be similar in almost all tumour types, but metastasis to different tissues seems to require distinct sets of regulators and/or an 'educated' microenvironment which may facilitate the infiltration and colonization of tumour cells to specific tissues. Moreover, interactions of tumour cells with stromal cells, endothelial cells, and immune cells that they encounter will also aid them to gain survival advantages, evade immune surveillance, and adapt to the new host microenvironment. Due to the high correlation between tumour metastasis and survival rate of patients, a deeper understanding of the molecular participants and processes involved in metastasis could pave the way towards novel, more effective and targeted approaches to prevent and treat tumour metastasis. In this review, we provide an update on the regulation networks orchestrated by the dominant regulators of different stages throughout the metastatic process including, but not limited to, epithelial-mesenchymal transition in local invasion, resistance to anoikis during migration, and colonization of different distant sites. We also put forward some suggestions and problems concerning the treatment of tumour metastasis that should be solved and/or improved for better therapies in the near future. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Neoplasm Metastasis/genetics , Neoplasms/genetics , Receptors, Notch/genetics , Signal Transduction , Transforming Growth Factor beta/genetics , Wnt Signaling Pathway/genetics , Anoikis/genetics , Cell Movement , Epithelial-Mesenchymal Transition , Humans , Models, Biological , Neoplasms/pathology , Stromal Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...