Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Leukemia ; 37(2): 441-452, 2023 02.
Article in English | MEDLINE | ID: mdl-36446947

ABSTRACT

DLBCL is the most common lymphoma with high tumor heterogeneity. Treatment refractoriness and relapse from R-CHOP therapy in patients remain a clinical problem. Activation of the non-canonical NF-κB pathway is associated with R-CHOP resistance. However, downstream targets of non-canonical NF-κB mediating R-CHOP-induced resistance remains uncharacterized. Here, we identify the common mechanisms underlying both intrinsic and acquired resistance that are induced by doxorubicin, the main cytotoxic component of R-CHOP. We performed global transcriptomic analysis of (1) a panel of resistant versus sensitive and (2) isogenic acquired doxorubicin-resistant DLBCL cell lines following short and chronic exposure to doxorubicin respectively. Doxorubicin-induced stress in resistant cells activates a distinct transcriptional signature that is enriched in metabolic reprogramming and oncogenic signalling. Selective and sustained activation of non-canonical NF-κB signalling in these resistant cells exacerbated their survival by augmenting glycolysis. In response to doxorubicin, p52-RelB complexes transcriptionally activated multiple glycolytic regulators with prognostic significance through increased recruitment at their gene promoters. Targeting p52-RelB and their targets in resistant cells increased doxorubicin sensitivity in vitro and in vivo. Collectively, our study uncovered novel molecular drivers of doxorubicin-induced resistance that are regulated by non-canonical NF-κB pathway. We reveal new avenues of therapeutic targeting for R-CHOP-treated refractory/relapsed DLBCL patients.


Subject(s)
Antineoplastic Agents , Lymphoma, Large B-Cell, Diffuse , Humans , NF-kappa B/metabolism , Neoplasm Recurrence, Local/drug therapy , Signal Transduction , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Antineoplastic Agents/therapeutic use , Rituximab/pharmacology , Rituximab/therapeutic use , Cyclophosphamide/therapeutic use , Vincristine/pharmacology , Vincristine/therapeutic use , Prednisone/therapeutic use , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
2.
Life Sci Alliance ; 4(7)2021 07.
Article in English | MEDLINE | ID: mdl-34117091

ABSTRACT

WBP2 is an emerging oncoprotein with diverse functions in breast tumorigenesis via regulating Wnt, epidermal growth factor receptor, estrogen receptor, and Hippo. Recently, evidence shows that WBP2 is tightly regulated by the components of the miRNA biogenesis machinery such as DGCR8 and Dicer via producing both WBP2's 3'UTR and coding DNA sequence-targeting miRNAs. This led us to hypothesize that WBP2 could provide a feedback loop to the biogenesis of its key upstream regulators by regulating the microprocessor complex activity. Indeed, WBP2 suppressed microprocessor activity by blocking the processing of pri-miRNAs to pre-miRNAs. WBP2 negatively regulated the assembly of the microprocessor complex via physical interactions with its components. Meta-analyses suggest that microprocessor complex components, in particular DGCR8, DDX5, and DEAD-Box Helicase17 (DDX17), have tumor-suppressive properties. 2D and 3D in vitro proliferation assays revealed that WBP2 blocked the tumor-suppressive properties of DGCR8, a key component of the microprocessor complex. In conclusion, WBP2 is a novel regulator of miRNA biogenesis that is a known dysregulated pathway in breast tumorigenesis. The reregulation of miRNA biogenesis machinery via targeting WBP2 protein may have implications in breast cancer therapy.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/biosynthesis , Trans-Activators/metabolism , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , DEAD-box RNA Helicases/metabolism , Female , Humans , MicroRNAs/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism , Trans-Activators/physiology
3.
Cell Death Dis ; 11(8): 669, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32820148

ABSTRACT

WBP2 transcription coactivator is an emerging oncoprotein and a key node of convergence between EGF and Wnt signaling pathways. Understanding how WBP2 is regulated has important implications for cancer therapy. WBP2 is tightly controlled by post-translational modifications, including phosphorylation and ubiquitination, leading to changes in subcellular localization, protein-protein interactions, and protein turnover. As the function of WBP2 is intricately linked to YAP and TAZ, we hypothesize that WBP2 is negatively regulated by the Hippo tumor suppressor pathway. Indeed, MST is demonstrated to negatively regulate WBP2 expression in a kinase-dependent but LATS-independent manner. This was observed in the majority of the breast cancer cell lines tested. The effect of MST was enhanced by SAV and concomitant with the inhibition of the transcription co-activation, in vitro and in vivo tumorigenesis activities of WBP2, resulting in good prognosis in xenografts. Downregulation of WBP2 by MST involved miRNA but not proteasomal or lysosomal degradation. Our data support the existence of a novel MST-Dicer signaling axis, which in turn regulates both WBP2 CDS- and UTR-targeting miRNAs expression, including miR-23a. MiR-23a targets the 3'UTR of WBP2 mRNA directly. Significant inverse relationships between WBP2 and MST or miR23a expression levels in clinical specimens were observed. In conclusion, WBP2 is a target of the Hippo/MST kinase; MST is identified as yet another rheostat in the regulation of WBP2 and its oncogenic function. The findings have implications in targeted therapeutics and precision medicine for breast cancer.


Subject(s)
Breast Neoplasms/metabolism , DEAD-box RNA Helicases/metabolism , Ribonuclease III/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Hippo Signaling Pathway , Humans , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/physiology , MCF-7 Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Oncogene Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Ribonuclease III/genetics , Signal Transduction/genetics , Trans-Activators/physiology , Transcription Factors/metabolism , Wnt Signaling Pathway
4.
J Biol Chem ; 293(52): 20014-20028, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30442712

ABSTRACT

The transcriptional coactivator WW domain-binding protein 2 (WBP2) is an emerging oncogene and serves as a node between the signaling protein Wnt and other signaling molecules and pathways, including epidermal growth factor receptor, estrogen receptor/progesterone receptor, and the Hippo pathway. The upstream regulation of WBP2 is well-studied, but its downstream activity remains unclear. Here, we elucidated WBP2's role in triple-negative breast cancer (TNBC), in which Wnt signaling is predominantly activated. Using RNAi coupled with RNA-Seq and MS analyses to identify Wnt/WBP2- and WBP2-dependent targets in MDA-MB-231 TNBC cells, we found that WBP2 is required for the expression of a core set of genes in Wnt signaling. These included AXIN2, which was essential for Wnt/WBP2-mediated breast cancer growth and migration. WBP2 also regulated a much larger set of genes and proteins independently of Wnt, revealing that WBP2 primes cells to Wnt activity by up-regulating G protein pathway suppressor 1 (GPS1) and TRAF2- and NCK-interacting kinase (TNIK). GPS1 activated the c-Jun N-terminal kinase (JNK)/Jun pathway, resulting in a positive feedback loop with TNIK that mediated Wnt-induced AXIN2 expression. WBP2 promoted TNBC growth by integrating JNK with Wnt signaling, and its expression profoundly influenced the sensitivity of TNBC to JNK/TNIK inhibitors. In conclusion, WBP2 links JNK to Wnt signaling in TNBC. GPS1 and TNIK are constituents of a WBP2-initiated cascade that primes responses to Wnt ligands and are also important for TNBC biology. We propose that WBP2 is a potential drug target for JNK/TNIK-based precision medicine for managing TNBC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neoplasm Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MCF-7 Cells , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Trans-Activators , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
5.
Cancer Res ; 76(21): 6278-6289, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27578003

ABSTRACT

Cross-talk between the Hippo and Wnt pathways has been implicated recently in breast cancer development, but key intersections have yet to be fully defined. Here we report that WBP2, a transcription coactivator that binds the Hippo pathway transcription factor YAP/TAZ, contributes to Wnt signaling and breast cancer pathogenesis. Clinically, overexpression of WBP2 in breast cancer specimens correlated with malignant progression and poor patient survival. In breast cancer cells, nuclear entry and interaction of WBP2 with ß-catenin was stimulated by Wnt3A, thereby activating TCF-mediated transcription and driving malignant invasive character. Mechanistic investigations showed WBP2 levels were controlled by the E3 ligase ITCH, which bound and target WBP2 for ubiquitin-dependent proteasomal degradation. Accordingly, ITCH silencing could elevate WBP2 levels. Wnt signaling upregulated WBP2 by disrupting ITCH-WBP2 interactions via EGFR-mediated tyrosine phosphorylation of WBP2 and TAZ/YAP competitive binding. Conversely, ITCH-mediated downregulation of WBP2 inhibited TCF/ß-catenin transcription, in vitro transformation, and in vivo tumorigenesis. We identified somatic mutations in ITCH, which impaired its ability to degrade WBP2 and to block its function in cancer, even while retaining binding capacity to WBP2. Thus, the Wnt pathway appeared to engage WBP2 primarily by affecting its protein stability. Our findings show how WBP2/ITCH signaling functions to link the intricate Wnt and Hippo signaling networks in breast cancer. Cancer Res; 76(21); 6278-89. ©2016 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Breast Neoplasms/pathology , Nuclear Proteins/metabolism , Repressor Proteins/physiology , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/physiology , Wnt Signaling Pathway/physiology , Acyltransferases , Animals , Cell Cycle Proteins , Cell Line, Tumor , ErbB Receptors/physiology , Female , Humans , Mice , Proteasome Endopeptidase Complex/physiology , Trans-Activators , Tumor Suppressor Proteins/physiology , Wnt3A Protein/physiology
6.
Genes Chromosomes Cancer ; 50(10): 837-47, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21769958

ABSTRACT

Understanding the molecular etiology of cancer and increasing the number of drugs and their targets are critical to cancer management. In our attempt to unravel novel breast-cancer associated proteins, we previously conducted protein expression profiling of the MCF10AT model, which comprises a series of isogenic cell lines that mimic different stages of breast cancer progression. NRD1 expression was found to increase during breast cancer progression. Here, we attempted to confirm the relevance of NRD1 in clinical breast cancer and understand the functional role and mechanism of NRD1 in breast cancer cells. Immunohistochemistry data show that NRD1 expression was elevated in ductal carcinoma in situ and invasive ductal carcinomas compared with normal tissues in 30% of the 26 matched cases studied. Examination of NRD1 expression in tissue microarray comprising >100 carcinomas and subsequent correlation with clinical data revealed that NRD1 expression was significantly associated with tumor size, grade, and nodal status (P < 0.05). Silencing of NRD1 reduced MCF10CA1h and MDA-MD-231 breast-cancer-cell proliferation and growth. Probing the oncogenic EGF signaling pathways revealed that NRD1 knock down did not affect overall downstream tyrosine phosphorylation cascades including AKT and MAPK activation. Instead, silencing of NRD1 resulted in a reduction of overall cyclin D1 expression, a reduction of EGF-induced increase in cyclin D1 expression and an increase in apoptotic cell population compared with control cells.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Cyclin D1/genetics , Metalloproteases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Disease Progression , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Silencing/drug effects , Humans , Immunohistochemistry , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Tyrosine/metabolism , Up-Regulation
7.
FASEB J ; 25(9): 3004-18, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21642474

ABSTRACT

WW-binding protein 2 (WBP2) has been demonstrated in different studies to be a tyrosine kinase substrate, to activate estrogen receptor α (ERα)/progesterone receptor (PR) transcription, and to play a role in breast cancer. However, the role of WBP2 tyrosine phosphorylation in regulating ERα function and breast cancer biology is unknown. Here, we established WBP2 as a tyrosine phosphorylation target of estrogen signaling via EGFR crosstalk. Using dominant-negative, constitutively active mutants, RNAi, and pharmacological studies, we demonstrated that phosphorylation of WBP2 at Tyr192 and Tyr231 could be regulated by c-Src and c-Yes kinases. We further showed that abrogating WBP2 phosphorylation impaired >60% of ERα reporter activity, putatively by blocking nuclear entry of WBP2 and its interaction with ERα. Compared to vector control, overexpression of WBP2 and its phospho-mimic mutant in MCF7 cells resulted in larger tumors in mice, induced loss of cell-cell adhesion, and enhanced cell proliferation, anchorage-independent growth, migration, and invasion in both estrogen-dependent and -independent manners, events of which could be substantially abolished by overexpression of the phosphorylation-defective mutant. Hormone independence of cells expressing WBP2 phospho-mimic mutant was associated with heightened ERα and Wnt reporter activities. Wnt/ß-catenin inhibitor FH535 blocked phospho-WBP2-mediated cancer cell growth more pronouncedly than tamoxifen and fulvestrant, in part by reducing the expression of ERα. Wnt pathway is likely to be a critical component in WBP2-mediated breast cancer biology.


Subject(s)
Carrier Proteins/metabolism , Estrogen Receptor alpha/metabolism , Mammary Neoplasms, Animal/metabolism , Neoplasms, Experimental/metabolism , Tyrosine/metabolism , Wnt Proteins/metabolism , Animals , Antineoplastic Agents , Carrier Proteins/genetics , Cell Line , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/physiology , Genes, src , Humans , Mice , Mice, Nude , Mutation , Phosphorylation , Proto-Oncogene Proteins c-yes , Trans-Activators , Wnt Proteins/genetics
8.
J Proteome Res ; 9(9): 4767-78, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20812763

ABSTRACT

Cancer progression is governed by multifaceted interactions of cancer cells with their microenvironment and one of these ways is through secreted compounds. Substances released by gastric cancer cells have not being profiled in a proteome-wide manner. ITRAQ-based tandem mass spectrometry was employed to quantify proteins secreted by HFE145 normal, MKN7 well-differentiated, and MKN45 poorly differentiated gastric cancer cell lines. The expression levels of 237 proteins were found to be significantly different between normal and cancer cells. Further examination of 16 gastric cell lines and 115 clinical samples validated the up-regulation of CTSS expression in gastric cancer. Silencing CTSS expression suppressed the migration and invasion of gastric cancer cells in vitro. Subsequent secretomics revealed that CTSS silencing resulted in changes in expression levels of 197 proteins, one-third of which are implicated in cellular movement. Proteome-wide comparative secretomes of normal and gastric cancer cells were produced that constitute a useful resource for gastric cancer research. CTSS was demonstrated to play novel roles in gastric cancer cell migration and invasion, putatively via a network of proteins associated with cell migration, invasion, or metastasis. Cathepsin S is member of a large group of extracellular proteases, which are attractive drug targets. The implicated role of CTSS in gastric cancer metastasis provides an opportunity to test existing compounds against CTSS for adjuvant therapy and/or treatment of metastatic gastric cancers.


Subject(s)
Cathepsins/metabolism , Cell Movement/physiology , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Cathepsins/chemistry , Cell Line, Tumor , Humans , Isotope Labeling , Neoplasm Invasiveness , Neoplasm Proteins/chemistry , Proteomics/methods , Reproducibility of Results , Signal Transduction , Tandem Mass Spectrometry
9.
Cell Signal ; 22(3): 437-46, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19887107

ABSTRACT

In our previous study, Endofin was validated to be a novel tyrosine phosphorylation target downstream of EGFR. Here, we attempted to map the signaling events associated with Endofin following activation of EGFR with EGF. Tyrosine phosphorylation of endogenous Endofin peaked around 15 min and was modulated within 30 min of EGF treatment. Phosphatidylinositol 3-kinase (PI3K) activity and FYVE domain-mediated localization of Endofin to EEA1-marked endosomes were shown to be necessary for the tyrosine phosphorylation of Endofin. Tyrosine 515 was mapped to be a major phosphorylation site on Endofin but disruption of phosphorylation at Y515 neither affected Endofin's localization nor its co-localization with EGFR in the endosomes. Instead, abrogation of Y515 phosphorylation and mislocalization of Endofin were found to enhance the amplitude of the MAPK cascade, suggesting a possible role of Endofin in the modulation of MAPK pathway. Our study has identified a novel signaling cascade involving EGFR, PI3K, Endofin and MAPK in the EGFR signaling network.


Subject(s)
Endosomes/metabolism , Epidermal Growth Factor/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cell Line , ErbB Receptors/metabolism , Humans , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/genetics , Kinetics , MAP Kinase Signaling System , Mutagenesis, Site-Directed , Phosphorylation , Protein Structure, Tertiary , Serine Endopeptidases/analysis , Serine Endopeptidases/genetics , Tyrosine/metabolism
10.
Biochem J ; 403(1): 119-27, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17125467

ABSTRACT

Mitotic Aurora-A is an oncogene, which undergoes a cell-cycle-dependent regulation of both its synthesis and degradation. Overexpression of Aurora-A leads to aneuploidy and cellular transformation in cultured cells. It has been shown that the cell-cycle-dependent turnover of Aurora-A is mediated by Cdh1 (CDC20 homologue 1) through the anaphase-promoting complex/cyclosome (APC/C)-ubiquitin-proteasome pathway. We have described previously the identification of an Aurora-A kinase interacting protein, AURKAIP1 (formerly described as AIP), which is also involved in the destabilization of Aurora-A through the proteasome-dependent degradation pathway. In an attempt to investigate the mechanism of AURKAIP1-mediated Aurora-A degradation, we report here that AURKAIP1 targets Aurora-A for degradation in a proteasome-dependent but Ub (ubiquitin)-independent manner. AURKAIP1 inhibits polyubiquitination of Aurora-A. A non-interactive AURKAIP1 mutant that cannot destabilize Aurora-A restores ubiquitination of Aurora-A. An A-box mutant of Aurora-A, which cannot be targeted for proteasome-dependent degradation by Cdh1, can still be degraded by AURKAIP1. Inhibition of cellular ubiquitination either by expression of dominant negative Ub mutants or by studies in ts-20 (temperature sensitive-20) CHO (Chinese-hamster ovary) cell line lacking the E1 Ub activating enzyme at the restrictive temperature, cannot abolish AURKAIP1-mediated degradation of Aurora-A. AURKAIP1 specifically decreases the stability of Aurora-A in ts-20 CHO cells at the restrictive temperature, while cyclinB1 and p21 are not affected. This demonstrates that there exists an Ub-independent alternative pathway for Aurora-A degradation and AURKAIP1 promotes Aurora-A degradation through this Ub-independent yet proteasome-dependent pathway.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Ubiquitin/metabolism , Amino Acid Substitution , Animals , Aurora Kinase A , Aurora Kinases , Cell Cycle , Flow Cytometry , Intracellular Signaling Peptides and Proteins , Kinetics , Mice , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Recombinant Proteins/metabolism , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...