Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Metabolism ; 151: 155746, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38016540

ABSTRACT

BACKGROUND: Multinucleation is a hallmark of osteoclast formation and has a unique ability to resorb bone matrix. During osteoclast differentiation, the cytoskeleton reorganization results in the generation of actin belts and eventual bone resorption. Tetraspanins are involved in adhesion, migration and fusion in various cells. However, its function in osteoclast is still unclear. In this study, we identified Tm4sf19, a member of the tetraspanin family, as a regulator of osteoclast function. MATERIALS AND METHODS: We investigate the effect of Tm4sf19 deficiency on osteoclast differentiation using bone marrow-derived macrophages obtained from wild type (WT), Tm4sf19 knockout (KO) and Tm4sf19 LELΔ mice lacking the large extracellular loop (LEL). We analyzed bone mass of young and aged WT, KO and LELΔ mice by µCT analysis. The effects of Tm4sf19 LEL-Fc fusion protein were accessed in osteoclast differentiation and osteoporosis animal model. RESULTS: We found that deficiency of Tm4sf19 inhibited osteoclast function and LEL of Tm4sf19 was responsible for its function in osteoclasts in vitro. KO and LELΔ mice exhibited higher trabecular bone mass compared to WT mice. We found that Tm4sf19 interacts with integrin αvß3 through LEL, and that this binding is important for cytoskeletal rearrangements in osteoclast by regulating signaling downstream of integrin αvß3. Treatment with LEL-Fc fusion protein inhibited osteoclast function in vitro and administration of LEL-Fc prevented bone loss in an osteoporosis mouse model in vivo. CONCLUSION: We suggest that Tm4sf19 regulates osteoclast function and that LEL-Fc may be a promising drug to target bone destructive diseases caused by osteoclast hyper-differentiation.


Subject(s)
Bone Diseases , Bone Resorption , Osteoporosis , Tetraspanins , Animals , Mice , Bone Resorption/genetics , Bone Resorption/metabolism , Cell Differentiation , Integrin alphaVbeta3/metabolism , Osteoclasts , Osteoporosis/genetics , Osteoporosis/metabolism , Tetraspanins/genetics , Tetraspanins/metabolism
2.
Cell Death Dis ; 14(2): 93, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36765032

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the most lethal type of cancer and the third leading cause of cancer death with the lowest 5-year survival rate. Heterogeneity, difficulty in diagnosis, and rapid metastatic progression are the causes of high mortality in pancreatic cancer. Recent studies have shown that Protein arginine methyltransferase 5 (PRMT5) is overexpressed in pancreatic cancers, and these patients have a worse prognosis. Recently, PRMT5 as an anti-cancer target has gained considerable interest. In this study, we investigated whether inhibition of PRMT5 activity was synergistic with blockade of TGF-ß1 signaling, which plays an important role in the construction of the desmoplastic matrix in pancreatic cancer and induces therapeutic vulnerability. Compared with T1-44, a selective inhibitor of PRMT5 activity, the combination of T1-44 with the TGF-ß1 signaling inhibitor Vactosertib significantly reduced tumor size and surrounding tissue invasion and significantly improved long-term survival. RNA sequencing analysis of mouse tumors revealed that the combination of T1-44 and Vactosertib significantly altered the expression of genes involved in cancer progression, such as cell migration, extracellular matrix, and apoptotic processes. In particular, the expression of Btg2, known as a tumor suppressor factor in various cancers, was markedly induced by combination treatment. Ectopic overexpression of Btg2 inhibited the EMT response, blocking cell migration, and promoted cancer cell death. These data demonstrate that the combination therapy of T1-44 with Vactosertib is synergistic for pancreatic cancer, suggesting that this novel combination therapy has value in the treatment strategy of patients with pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Transforming Growth Factor beta1/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Enzyme Inhibitors/therapeutic use , Cell Line, Tumor , Pancreatic Neoplasms
3.
Nat Commun ; 13(1): 6274, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36307405

ABSTRACT

Although accumulating evidence indicates that alternative splicing is aberrantly altered in many cancers, the functional mechanism remains to be elucidated. Here, we show that epithelial and mesenchymal isoform switches of leucine-rich repeat Fli-I-interacting protein 2 (LRRFIP2) regulated by epithelial splicing regulatory protein 1 (ESRP1) correlate with metastatic potential of gastric cancer cells. We found that expression of the splicing variants of LRRFIP2 was closely correlated with that of ESRP1. Surprisingly, ectopic expression of the mesenchymal isoform of LRRFIP2 (variant 3) dramatically increased liver metastasis of gastric cancer cells, whereas deletion of exon 7 of LRRFIP2 by the CRISPR/Cas9 system caused an isoform switch, leading to marked suppression of liver metastasis. Mechanistically, the epithelial LRRFIP2 isoform (variant 2) inhibited the oncogenic function of coactivator-associated arginine methyltransferase 1 (CARM1) through interaction. Taken together, our data reveals a mechanism of LRRFIP2 isoform switches in gastric cancer with important implication for cancer metastasis.


Subject(s)
Liver Neoplasms , Stomach Neoplasms , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alternative Splicing , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Liver Neoplasms/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Stomach Neoplasms/genetics , Transcription Factors/metabolism , Neoplasm Metastasis
4.
Cell Death Dis ; 13(2): 169, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194034

ABSTRACT

Despite favorable responses to initial chemotherapy, drug resistance is a major cause limiting chemotherapeutic efficacy in many advanced cancers. However, mechanisms that drive drug-specific resistance in chemotherapy for patients with advanced cancers are still unclear. Here, we report a unique role of death-associated protein kinase-related apoptosis-inducing kinase 1 (DRAK1) associated with paclitaxel resistance in cervical cancer cells. Interestingly, DRAK1 protein level was markedly decreased in paclitaxel-resistant cervical cancer cells without affecting its mRNA expression, which resulted in an increase in tumor necrosis factor receptor-associated factor 6 (TRAF6) expression, as well as an activation of TRAF6-mediated nuclear factor-kappa B (NF-κB) signaling cascade, thereby promoting tumor progression. DRAK1 depletion markedly increased the chemotherapeutic IC50 values of paclitaxel in cervical cancer cells. Ectopic expression of DRAK1 inhibited growth of paclitaxel-resistant cervical cancer cells in vitro and in vivo. Furthermore, DRAK1 was markedly underexpressed in chemoresistant cervical cancer patient tissues compared with chemosensitive samples. We found that DRAK1 protein was destabilized through K48-linked polyubiquitination promoted by the Cullin scaffold protein 3 (CUL3) / speckle-type POZ (poxvirus and zinc finger protein) protein (SPOP) E3 ubiquitin ligase in paclitaxel-resistant cells. Collectively, these findings suggest that DRAK1 may serve as a potential predictive biomarker for overcoming paclitaxel resistance in cervical cancer.


Subject(s)
Apoptosis Regulatory Proteins , Cullin Proteins , Nuclear Proteins , Protein Serine-Threonine Kinases , Repressor Proteins , Ubiquitin-Protein Ligases , Uterine Cervical Neoplasms , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism , Female , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Paclitaxel/therapeutic use , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics
5.
Cancers (Basel) ; 13(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34572878

ABSTRACT

Few studies have examined the role of BAG2 in malignancies. We investigated the prognostic value of BAG2-expression in cancer-associated fibroblasts (CAFs) and tumor cells in predicting metastasis-free survival in patients with breast cancer. Tissue-microarray was constructed using human breast cancer tissues obtained by surgical resection between 1992 and 2015. BAG2 expression was evaluated by immunohistochemistry in CAFs or the tumor cells. BAG2 expression in the CAFs and cytoplasm of tumor cells was classified as positive and negative, and low and high, respectively. BAG2-CAF was evaluated in 310 patients and was positive in 67 (21.6%) patients. Kaplan-Meier plots showed that distant metastasis-free survival (DMFS) was lesser in patients with BAG2(+) CAF than in patients with BAG2(-) CAF (p = 0.039). Additionally, we classified the 310 patients into two groups: 109 in either BAG2-high or BAG2(+) CAF and 201 in BAG2-low and BAG2(-) CAF. DMFS was significantly reduced in patients with either BAG2-high or BAG2(+) CAF than in the patients of the other group (p = 0.005). Multivariable analysis demonstrated that DMFS was prolonged in patients with BAG2(-) CAF or BAG2-low. Evaluation of BAG2 expression on both CAFs and tumor cells could help in determining the risk of metastasis in breast cancer.

6.
Cell Death Dis ; 12(2): 159, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558527

ABSTRACT

Although tetraarsenic hexoxide is known to exert an anti-tumor effect by inducing apoptosis in various cancer cells, its effect on other forms of regulated cell death remains unclear. Here, we show that tetraarsenic hexoxide induces the pyroptotic cell death through activation of mitochondrial reactive oxygen species (ROS)-mediated caspase-3/gasdermin E (GSDME) pathway, thereby suppressing tumor growth and metastasis of triple-negative breast cancer (TNBC) cells. Interestingly, tetraarsenic hexoxide-treated TNBC cells exhibited specific pyroptotic characteristics, including cell swelling, balloon-like bubbling, and LDH releases through pore formation in the plasma membrane, eventually suppressing tumor formation and lung metastasis of TNBC cells. Mechanistically, tetraarsenic hexoxide markedly enhanced the production of mitochondrial ROS by inhibiting phosphorylation of mitochondrial STAT3, subsequently inducing caspase-3-dependent cleavage of GSDME, which consequently promoted pyroptotic cell death in TNBC cells. Collectively, our findings highlight tetraarsenic hexoxide-induced pyroptosis as a new therapeutic strategy that may inhibit cancer progression of TNBC cells.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenic Trioxide/pharmacology , Caspase 3/metabolism , Mitochondria/drug effects , Pyroptosis/drug effects , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Caspase 3/genetics , Cell Line, Tumor , Enzyme Activation , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Phosphorylation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
7.
Nat Commun ; 11(1): 4681, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943626

ABSTRACT

Although advanced lipidomics technology facilitates quantitation of intracellular lipid components, little is known about the regulation of lipid metabolism in cancer cells. Here, we show that disruption of the Gdpd3 gene encoding a lysophospholipase D enzyme significantly decreased self-renewal capacity in murine chronic myelogenous leukaemia (CML) stem cells in vivo. Sophisticated lipidomics analyses revealed that Gdpd3 deficiency reduced levels of certain lysophosphatidic acids (LPAs) and lipid mediators in CML cells. Loss of Gdpd3 also activated AKT/mTORC1 signalling and cell cycle progression while suppressing Foxo3a/ß-catenin interaction within CML stem cell nuclei. Strikingly, CML stem cells carrying a hypomorphic mutation of Lgr4/Gpr48, which encodes a leucine-rich repeat (LRR)-containing G-protein coupled receptor (GPCR) acting downstream of Gdpd3, displayed inadequate disease-initiating capacity in vivo. Our data showing that lysophospholipid metabolism is required for CML stem cell maintenance in vivo establish a new, biologically significant mechanism of cancer recurrence that is independent of oncogene addiction.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Phosphoric Diester Hydrolases/metabolism , Stem Cells/metabolism , Animals , Disease Models, Animal , Female , Forkhead Box Protein O3/metabolism , Lysophospholipids/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neoplasm Recurrence, Local/metabolism , Phosphoric Diester Hydrolases/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , beta Catenin/metabolism
8.
Cancer Res ; 80(12): 2537-2549, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32265222

ABSTRACT

The adaptor protein TNF receptor-associated factor 6 (TRAF6) is a key mediator in inflammation. However, the molecular mechanisms controlling its activity and stability in cancer progression remain unclear. Here we show that death-associated protein kinase-related apoptosis-inducing kinase 1 (DRAK1) inhibits the proinflammatory signaling pathway by targeting TRAF6 for degradation, thereby suppressing inflammatory signaling-mediated tumor growth and metastasis in advanced cervical cancer cells. DRAK1 bound directly to the TRAF domain of TRAF6, preventing its autoubiquitination by interfering with homo-oligomerization, eventually leading to autophagy-mediated degradation of TRAF6. Depletion of DRAK1 in cervical cancer cells resulted in markedly increased levels of TRAF6 protein, promoting activation of the IL1ß signaling-associated pathway and proinflammatory cytokine production. DRAK1 was specifically underexpressed in metastatic cervical cancers and inversely correlated with TRAF6 expression in mouse xenograft model tumor tissues and human cervical tumor tissues. Collectively, our findings highlight DRAK1 as a novel antagonist of inflammation targeting TRAF6 for degradation that limits inflammatory signaling-mediated progression of advanced cervical cancer. SIGNIFICANCE: Serine/threonine kinase DRAK1 serves a unique role as a novel negative regulator of the inflammatory signaling mediator TRAF6 in cervical cancer progression.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Uterine Cervical Neoplasms/pathology , Animals , Cell Line, Tumor , Disease Progression , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Mice , Neoplasm Staging , Protein Binding/immunology , Protein Domains , Protein Multimerization/immunology , Protein Stability , Proteolysis , Signal Transduction/immunology , Tissue Array Analysis , Ubiquitination/immunology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology , Xenograft Model Antitumor Assays
9.
Sci Rep ; 10(1): 2935, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32076068

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies. TGF-ß is strongly expressed in both the epithelial and stromal compartments of PDAC, and dysregulation of TGF-ß signalling is a frequent molecular disturbance in PDAC progression and metastasis. In this study, we investigated whether blockade of TGF-ß signalling synergizes with nal-IRI/5-FU/LV, a chemotherapy regimen for malignant pancreatic cancer, in an orthotopic pancreatic tumour mouse model. Compared to nal-IRI/5-FU/LV treatment, combining nal-IRI/5-FU/LV with vactosertib, a TGF-ß signalling inhibitor, significantly improved long-term survival rates and effectively suppressed invasion to surrounding tissues. Through RNA-sequencing analysis, we identified that the combination treatment results in robust abrogation of tumour-promoting gene signatures and positive enrichment of tumour-suppressing and apoptotic gene signatures. Particularly, the expression of tumour-suppressing gene Ccdc80 was induced by vactosertib and further induced by vactosertib in combination with nal-IRI/5-FU/LV. Ectopic expression of CCDC80 suppressed migration and colony formation concomitant with decreased expression of epithelial-to-mesenchymal transition (EMT) markers in pancreatic cancer cells. Collectively, these results indicate that combination treatment of vactosertib with nal-IRI/5-FU/LV improves overall survival rates in a mouse model of pancreatic cancer by suppressing invasion through CCDC80. Therefore, combination therapy of nal-IRI/5-FU/LV with vactosertib could provide clinical benefits to pancreatic cancer patients.


Subject(s)
Fluorouracil/therapeutic use , Irinotecan/therapeutic use , Leucovorin/therapeutic use , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Irinotecan/pharmacology , Leucovorin/pharmacology , Liposomes , Mice, Inbred C57BL , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Survival Analysis , Transcriptome/genetics , Triazoles/pharmacology , Triazoles/therapeutic use , Tumor Stem Cell Assay , Up-Regulation/drug effects
10.
Nat Commun ; 10(1): 5805, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31862882

ABSTRACT

The development of triple-negative breast cancer (TNBC) negatively impacts both quality of life and survival in a high percentage of patients. Here, we show that RING finger protein 208 (RNF208) decreases the stability of soluble Vimentin protein through a polyubiquitin-mediated proteasomal degradation pathway, thereby suppressing metastasis of TNBC cells. RNF208 was significantly lower in TNBC than the luminal type, and low expression of RNF208 was strongly associated with poor clinical outcomes. Furthermore, RNF208 was induced by 17ß-estradiol (E2) treatment in an estrogen receptor alpha (ΕRα)-dependent manner. Overexpression of RNF208 suppresses tumor formation and lung metastasis of TNBC cells. Mechanistically, RNF208 specifically polyubiquitinated the Lys97 residue within the head domain of Vimentin through interaction with the Ser39 residue of phosphorylated Vimentin, which exists as a soluble form, eventually facilitating proteasomal degradation of Vimentin. Collectively, our findings define RNF208 as a negative regulator of soluble Vimentin and a prognostic biomarker for TNBC cells.


Subject(s)
Estradiol/metabolism , Lung Neoplasms/secondary , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Vimentin/metabolism , Animals , Breast/pathology , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Female , Humans , Lung/pathology , Mice , Prognosis , Protein Stability , Proteolysis , Survival Analysis , Tissue Array Analysis , Triple Negative Breast Neoplasms/mortality , Ubiquitination , Xenograft Model Antitumor Assays
11.
PLoS One ; 14(8): e0221721, 2019.
Article in English | MEDLINE | ID: mdl-31449546

ABSTRACT

BACKGROUND: A20 protein has ubiquitin-editing activities and acts as a key regulator of inflammation and immunity. Previously, our group showed that A20 promotes tumor metastasis through multi-monoubiquitylation of SNAIL1 in basal-like breast cancer. Here, we investigated survival outcomes in patients with breast cancer according to A20 expression. PATIENTS AND METHODS: We retrospectively collected tumor samples from patients with breast cancer. Immunohistochemistry (IHC) with an A20-specific antibody was performed, and survival outcomes were analyzed. RESULTS: A20 expression was evaluated in 442 patients. High A20 expression was associated with advanced anatomical stage and young age. High A20 expression showed significantly inferior recurrence-free-survival and overall-survival (P<0.001 and P<0.001, respectively). Multivariate analysis showed that A20 was an independent prognostic marker for RFS (HRs: 2.324, 95% CIs: 1.446-3.736) and OS (HRs: 2.629, 95% CIs: 1.585-4.361). In human epidermal growth factor receptor 2 (HER2)-positive and triple negative breast cancer (TNBC) subtypes, high A20 levels were associated with poor OS. CONCLUSION: We found that A20 expression is a poor prognostic marker in breast cancer. The prognostic impact of A20 was pronounced in aggressive tumors, such as HER2-positive and TNBC subtypes. Our findings suggested that A20 may be a valuable target in patients with aggressive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Adult , Aged , Aged, 80 and over , Confidence Intervals , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/pathology , Prognosis , Proportional Hazards Models , Survival Analysis , Young Adult
12.
Cancer Sci ; 110(2): 481-488, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30589983

ABSTRACT

Smad3, a major transcription factor in transforming growth factor-ß (TGF-ß) signaling, plays critical roles in both tumor-suppressive and pro-oncogenic functions. Upon TGF-ß stimulation, the C-terminal tail of Smad3 undergoes phosphorylation that is essential for canonical TGF-ß signaling. The Smad3 linker region contains serine/threonine phosphorylation sites and can be phosphorylated by intracellular kinases, such as the MAPK family, cyclin-dependent kinase (CDK) family and glycogen synthase kinase-3ß (GSK-3ß). Previous reports based on cell culture studies by us and others showed that mutation of Smad3 linker phosphorylation sites dramatically intensifies TGF-ß responses as well as growth-inhibitory function and epithelial-mesenchymal transition (EMT), suggesting that Smad3 linker phosphorylation suppresses TGF-ß transcriptional activities. However, recent discoveries of Smad3-interacting molecules that preferentially bind phosphorylated Smad3 linker serine/threonine residues have shown a multitude of signal transductions that either enhance or suppress TGF-ß responses associated with Smad3 turnover or cancer progression. This review aims at providing new insight into the perplexing mechanisms of TGF-ß signaling affected by Smad3 linker phosphorylation and further attempts to gain insight into elimination and protection of TGF-ß-mediated oncogenic and growth-suppressive signals, respectively.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/physiology , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Disease Progression , Humans
14.
J Cancer Prev ; 23(1): 1-9, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29629343

ABSTRACT

BACKGROUND: Smad3 linker phosphorylation plays essential roles in tumor progression and metastasis. We have previously reported that the mutation of Smad3 linker phosphorylation sites (Smad3-Erk/Pro-directed kinase site mutant constructs [EPSM]) markedly reduced the tumor progression while increasing the lung metastasis in breast cancer. METHODS: We performed high-throughput RNA-Sequencing of the human prostate cancer cell lines infected with adenoviral Smad3-EPSM to identify the genes regulated by Smad3-EPSM. RESULTS: In this study, we identified genes which are differentially regulated in the presence of Smad3-EPSM. We first confirmed that Smad3-EPSM strongly enhanced a capability of cell motility and invasiveness as well as the expression of epithelial-mesenchymal transition marker genes, CDH2, SNAI1, and ZEB1 in response to TGF-ß1 in human pancreatic and prostate cancer cell lines. We identified GADD45B, CTGF, and JUNB genes in the expression profiles associated with cell motility and invasiveness induced by the Smad3-EPSM. CONCLUSIONS: These results suggested that inhibition of Smad3 linker phosphorylation may enhance cell motility and invasiveness by inducing expression of GADD45B, CTGF, and JUNB genes in various cancers.

15.
Cell Rep ; 21(10): 2952-2964, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212038

ABSTRACT

Triple-negative breast cancer (TNBC) is considered incurable with currently available treatments, highlighting the need for therapeutic targets and predictive biomarkers. Here, we report a unique role for Bcl-2-associated athanogene 2 (BAG2), which is significantly overexpressed in TNBC, in regulating the dual functions of cathepsin B as either a pro- or anti-oncogenic enzyme. Silencing BAG2 suppresses tumorigenesis and lung metastasis and induces apoptosis by increasing the intracellular mature form of cathepsin B, whereas BAG2 expression induces metastasis by blocking the auto-cleavage processing of pro-cathepsin B via interaction with the propeptide region. BAG2 regulates pro-cathepsin B/annexin II complex formation and facilitates the trafficking of pro-cathespin-B-containing TGN38-positive vesicles toward the cell periphery, leading to the secretion of pro-cathepsin B, which induces metastasis. Collectively, our results uncover BAG2 as a regulator of the oncogenic function of pro-cathepsin B and a potential diagnostic and therapeutic target that may reduce the burden of metastatic breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cathepsin B/metabolism , Molecular Chaperones/metabolism , Triple Negative Breast Neoplasms/metabolism , Apoptosis/genetics , Apoptosis/physiology , Cathepsin B/genetics , Cell Line, Tumor , Female , Humans , Molecular Chaperones/genetics , Triple Negative Breast Neoplasms/genetics
16.
Biochem Biophys Res Commun ; 494(3-4): 706-713, 2017 12 16.
Article in English | MEDLINE | ID: mdl-29097203

ABSTRACT

Smad3 linker phosphorylation is a candidate target for several kinases that play important roles in cancer cell initiation, proliferation and progression. Also, Smad3 is an essential intracellular mediator of TGF-ß1-induced transcriptional responses during carcinogenesis. Therefore, it is highly advantageous to identify and develop inhibitors targeting Smad3 linker phosphorylation for the treatment of cancers. Galangin (3,5,7-trihydroxyflavone) has been known to be an active flavonoid showing a cytotoxic effect on several cancer cells. However, the mechanism of action of galangin in various cancers remains unclear, and there has been no report concerning regulation of Smad3 phosphorylation by galangin. In the present study, we show that galangin significantly induced apoptosis and inhibited cell proliferation in the presence of TGF-ß1 in both human prostate and pancreatic cancer cell lines. Particularly, galangin effectively inhibits phosphorylation of the Thr-179 site at Smad3 linker region through suppression of CDK4 phosphorylation. Thus, galangin can be a promising candidate as a selective inhibitor to suppress phosphorylation of Smad3 linker region.


Subject(s)
Cell Proliferation/drug effects , Flavonoids/administration & dosage , Neoplasms/drug therapy , Neoplasms/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Humans , Neoplasms/pathology , Phosphorylation/drug effects , Protein Binding , Threonine/metabolism , Treatment Outcome
17.
Nat Cell Biol ; 19(10): 1260-1273, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28892081

ABSTRACT

Although the ubiquitin-editing enzyme A20 is a key player in inflammation and autoimmunity, its role in cancer metastasis remains unknown. Here we show that A20 monoubiquitylates Snail1 at three lysine residues and thereby promotes metastasis of aggressive basal-like breast cancers. A20 is significantly upregulated in human basal-like breast cancers and its expression level is inversely correlated with metastasis-free patient survival. A20 facilitates TGF-ß1-induced epithelial-mesenchymal transition (EMT) of breast cancer cells through multi-monoubiquitylation of Snail1. Monoubiquitylated Snail1 has reduced affinity for glycogen synthase kinase 3ß (GSK3ß), and is thus stabilized in the nucleus through decreased phosphorylation. Knockdown of A20 or overexpression of Snail1 with mutation of the monoubiquitylated lysine residues into arginine abolishes lung metastasis in mouse xenograft and orthotopic breast cancer models, indicating that A20 and monoubiquitylated Snail1 are required for metastasis. Our findings uncover an essential role of the A20-Snail1 axis in TGF-ß1-induced EMT and metastasis of basal-like breast cancers.


Subject(s)
Breast Neoplasms/enzymology , Cell Movement , Lung Neoplasms/enzymology , Snail Family Transcription Factors/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Ubiquitination , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement/drug effects , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lysine , MCF-7 Cells , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Phosphorylation , Protein Stability , RNA Interference , Signal Transduction , Snail Family Transcription Factors/genetics , Time Factors , Transfection , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination/drug effects
18.
Cancer Sci ; 107(2): 140-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26583567

ABSTRACT

Recent strategies for treating CML patients have focused on investigating new combinations of tyrosine kinase inhibitors (TKIs) as well as identifying novel translational research agents that can eradicate CML leukemia-initiating cells (CML-LICs). However, little is known about the therapeutic benefits such CML-LIC targeting therapies might bring to CML patients. In this study, we investigated the therapeutic potential of EW-7197, an orally bioavailable transforming growth factor-ß signaling inhibitor which has recently been approved as an Investigational New Drug (NIH, USA), to suppress CML-LICs in vivo. Compared to TKI treatment alone, administration of TKI plus EW-7197 to CML-affected mice significantly delayed disease relapse and prolonged survival. Notably, combined treatment with EW-7197 plus TKI was effective in eliminating CML-LICs even if they expressed the TKI-resistant T315I mutant BCR-ABL1 oncogene. Collectively, these results indicate that EW-7197 may be a promising candidate for a new therapeutic that can greatly benefit CML patients by working in combination with TKIs to eradicate CML-LICs.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Triazoles/pharmacology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Imidazoles/administration & dosage , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridazines/administration & dosage , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transfection , Transforming Growth Factor beta/antagonists & inhibitors
19.
Nat Commun ; 6: 8039, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26289811

ABSTRACT

Understanding the specific survival of the rare chronic myelogenous leukaemia (CML) stem cell population could provide a target for therapeutics aimed at eradicating these cells. However, little is known about how survival signalling is regulated in CML stem cells. In this study, we survey global metabolic differences between murine normal haematopoietic stem cells (HSCs) and CML stem cells using metabolomics techniques. Strikingly, we show that CML stem cells accumulate significantly higher levels of certain dipeptide species than normal HSCs. Once internalized, these dipeptide species activate amino-acid signalling via a pathway involving p38MAPK and the stemness transcription factor Smad3, which promotes CML stem cell maintenance. Importantly, pharmacological inhibition of dipeptide uptake inhibits CML stem cell activity in vivo. Our results demonstrate that dipeptide species support CML stem cell maintenance by activating p38MAPK-Smad3 signalling in vivo, and thus point towards a potential therapeutic target for CML treatment.


Subject(s)
Dipeptides/classification , Gene Expression Regulation, Neoplastic/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Neoplastic Stem Cells/metabolism , Smad3 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , DNA, Complementary , Dipeptides/metabolism , Female , Male , Mice , Mice, Transgenic , Retroviridae , Signal Transduction/physiology , Smad3 Protein/genetics , Symporters/genetics , Symporters/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
20.
Cancer Res ; 74(21): 6139-49, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205100

ABSTRACT

Smad3, a major intracellular mediator of TGFß signaling, functions as both a positive and negative regulator in carcinogenesis. In response to TGFß, the TGFß receptor phosphorylates serine residues at the Smad3 C-tail. Cancer cells often contain high levels of the MAPK and CDK activities, which can lead to the Smad3 linker region becoming highly phosphorylated. Here, we report, for the first time, that mutation of the Smad3 linker phosphorylation sites markedly inhibited primary tumor growth, but significantly increased lung metastasis of breast cancer cell lines. In contrast, mutation of the Smad3 C-tail phosphorylation sites had the opposite effect. We show that mutation of the Smad3 linker phosphorylation sites greatly intensifies all TGFß-induced responses, including growth arrest, apoptosis, reduction in the size of putative cancer stem cell population, epithelial-mesenchymal transition, and invasive activity. Moreover, all TGFß responses were completely lost on mutation of the Smad3 C-tail phosphorylation sites. Our results demonstrate a critical role of the counterbalance between the Smad3 C-tail and linker phosphorylation in tumorigenesis and metastasis. Our findings have important implications for therapeutic intervention of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Smad3 Protein/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Phosphorylation/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...