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










Publication year range
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.
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
3.
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
4.
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
5.
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
6.
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
8.
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.

9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...