Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
BMB Rep ; 57(6): 293-298, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835115

ABSTRACT

Microtubule acetylation has been shown to regulate actin filament dynamics by modulating signaling pathways that control actin organization, although the precise mechanisms remain unknown. In this study, we found that the downregulation of microtubule acetylation via the disruption ATAT1 (which encodes α-tubulin N-acetyltransferase 1) inhibited the expression of RhoA, a small GTPase involved in regulating the organization of actin filaments and the formation of stress fibers. Analysis of RHOA promoter and chromatin immunoprecipitation assays revealed that C/EBPß is a major regulator of RHOA expression. Interestingly, the majority of C/EBPß in ATAT1 knockout (KO) cells was found in the nucleus as a 27-kDa fragment (referred to as C/EBPßp27) lacking the N-terminus of C/EBPß. Overexpression of a gene encoding a C/EBPßp27-mimicking protein via an N-terminal deletion in C/EBPß led to competitive binding with wild-type C/EBPß at the C/EBPß binding site in the RHOA promoter, resulting in a significant decrease of RHOA expression. We also found that cathepsin L (CTSL), which is overexpressed in ATAT1 KO cells, is responsible for C/EBPßp27 formation in the nucleus. Treatment with a CTSL inhibitor led to the restoration of RHOA expression by downregulation of C/EBPßp27 and the invasive ability of ATAT1 KO MDA-MB-231 breast cancer cells. Collectively, our findings suggest that the downregulation of microtubule acetylation associated with ATAT1 deficiency suppresses RHOA expression by forming C/EBPßp27 in the nucleus through CTSL. We propose that CTSL and C/EBPßp27 may represent a novel therapeutic target for breast cancer treatment. [BMB Reports 2024; 57(6): 293-298].


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Down-Regulation , rhoA GTP-Binding Protein , Humans , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , Down-Regulation/genetics , Acetyltransferases/metabolism , Acetyltransferases/genetics , Promoter Regions, Genetic/genetics , Acetylation , Cathepsin L/metabolism , Cathepsin L/genetics , Microtubules/metabolism , Cell Line, Tumor
2.
bioRxiv ; 2023 May 20.
Article in English | MEDLINE | ID: mdl-37292765

ABSTRACT

Overexpression of repetitive elements is an emerging hallmark of human cancers 1 . Diverse repeats can mimic viruses by replicating within the cancer genome through retrotransposition, or presenting pathogen-associated molecular patterns (PAMPs) to the pattern recognition receptors (PRRs) of the innate immune system 2-5 . Yet, how specific repeats affect tumor evolution and shape the tumor immune microenvironment (TME) in a pro- or anti-tumorigenic manner remains poorly defined. Here, we integrate whole genome and total transcriptome data from a unique autopsy cohort of multiregional samples collected in pancreatic ductal adenocarcinoma (PDAC) patients, into a comprehensive evolutionary analysis. We find that more recently evolved S hort I nterspersed N uclear E lements (SINE), a family of retrotransposable repeats, are more likely to form immunostimulatory double-strand RNAs (dsRNAs). Consequently, younger SINEs are strongly co-regulated with RIG-I like receptor associated type-I interferon genes but anti-correlated with pro-tumorigenic macrophage infiltration. We discover that immunostimulatory SINE expression in tumors is regulated by either L ong I nterspersed N uclear E lements 1 (LINE1/L1) mobility or ADAR1 activity in a TP53 mutation dependent manner. Moreover, L1 retrotransposition activity tracks with tumor evolution and is associated with TP53 mutation status. Altogether, our results suggest pancreatic tumors actively evolve to modulate immunogenic SINE stress and induce pro-tumorigenic inflammation. Our integrative, evolutionary analysis therefore illustrates, for the first time, how dark matter genomic repeats enable tumors to co-evolve with the TME by actively regulating viral mimicry to their selective advantage.

3.
J Clin Invest ; 132(16)2022 08 15.
Article in English | MEDLINE | ID: mdl-35708912

ABSTRACT

Aberrant expression of viral-like repeat elements is a common feature of epithelial cancers, and the substantial diversity of repeat species provides a distinct view of the cancer transcriptome. Repeatome profiling across ovarian, pancreatic, and colorectal cell lines identifies distinct clustering independent of tissue origin that is seen with coding gene analysis. Deeper analysis of ovarian cancer cell lines demonstrated that human satellite II (HSATII) satellite repeat expression was highly associated with epithelial-mesenchymal transition (EMT) and anticorrelated with IFN-response genes indicative of a more aggressive phenotype. SATII expression - and its correlation with EMT and anticorrelation with IFN-response genes - was also found in ovarian cancer RNA-Seq data and was associated with significantly shorter survival in a second independent cohort of patients with ovarian cancer. Repeat RNAs were enriched in tumor-derived extracellular vesicles capable of stimulating monocyte-derived macrophages, demonstrating a mechanism that alters the tumor microenvironment with these viral-like sequences. Targeting of HSATII with antisense locked nucleic acids stimulated IFN response and induced MHC I expression in ovarian cancer cell lines, highlighting a potential strategy of modulating the repeatome to reestablish antitumor cell immune surveillance.


Subject(s)
Ovarian Neoplasms , RNA, Satellite , Carcinoma, Ovarian Epithelial/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Ovarian Neoplasms/genetics , Phenotype , RNA , Tumor Microenvironment/genetics
4.
Cancer Discov ; 12(6): 1462-1481, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35320348

ABSTRACT

Altered RNA expression of repetitive sequences and retrotransposition are frequently seen in colorectal cancer, implicating a functional importance of repeat activity in cancer progression. We show the nucleoside reverse transcriptase inhibitor 3TC targets activities of these repeat elements in colorectal cancer preclinical models with a preferential effect in p53-mutant cell lines linked with direct binding of p53 to repeat elements. We translate these findings to a human phase II trial of single-agent 3TC treatment in metastatic colorectal cancer with demonstration of clinical benefit in 9 of 32 patients. Analysis of 3TC effects on colorectal cancer tumorspheres demonstrates accumulation of immunogenic RNA:DNA hybrids linked with induction of interferon response genes and DNA damage response. Epigenetic and DNA-damaging agents induce repeat RNAs and have enhanced cytotoxicity with 3TC. These findings identify a vulnerability in colorectal cancer by targeting the viral mimicry of repeat elements. SIGNIFICANCE: Colorectal cancers express abundant repeat elements that have a viral-like life cycle that can be therapeutically targeted with nucleoside reverse transcriptase inhibitors (NRTI) commonly used for viral diseases. NRTIs induce DNA damage and interferon response that provide a new anticancer therapeutic strategy. This article is highlighted in the In This Issue feature, p. 1397.


Subject(s)
Colorectal Neoplasms , RNA-Directed DNA Polymerase , Animals , Antiviral Agents , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA , Humans , Interferons/metabolism , Lamivudine , Life Cycle Stages , RNA , RNA-Directed DNA Polymerase/metabolism , Tumor Suppressor Protein p53/genetics
5.
BMB Rep ; 55(4): 192-197, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35321783

ABSTRACT

Cell signals for growth factors depend on the mechanical properties of the extracellular matrix (ECM) surrounding the cells. Microtubule acetylation is involved in the transforming growth factor (TGF)-ß-induced myofibroblast differentiation in the soft ECM. However, the mechanism of activation of α-tubulin acetyltransferase 1 (α-TAT1), a major α-tubulin acetyltransferase, in the soft ECM is not well defined. Here, we found that casein kinase 2 (CK2) is required for the TGF-ß-induced activation of α-TAT1 that promotes microtubule acetylation in the soft matrix. Genetic mutation and pharmacological inhibition of CK2 catalytic activity specifically reduced microtubule acetylation in the cells cultured on a soft matrix rather than those cultured on a stiff matrix. Immunoprecipitation analysis showed that CK2α, a catalytic subunit of CK2, directly bound to the C-terminal domain of α-TAT1, and this interaction was more prominent in the cells cultured on the soft matrix. Moreover, the substitution of alanine with serine, the 236th amino acid located at the C-terminus, which contains the CK2-binding site of α-TAT1, significantly abrogated the TGF-ß-induced microtubule acetylation in the soft matrix, indicating that the successful binding of CK2 and the C-terminus of α-TAT1 led to the phosphorylation of serine at the 236th position of amino acids in α-TAT1 and regulation of its catalytic activity. Taken together, our findings provide novel insights into the molecular mechanisms underlying the TGF-ß-induced activation of α-TAT1 in a soft matrix. [BMB Reports 2022; 55(4): 192-197].


Subject(s)
Casein Kinase II , Fibroblasts , Acetyltransferases , Casein Kinase II/metabolism , Fibroblasts/metabolism , Phosphorylation , Serine/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
6.
Biomedicines ; 8(9)2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32917017

ABSTRACT

Microtubules are one of the major targets for anticancer drugs because of their role in cell proliferation and migration. However, as anticancer drugs targeting microtubules have side effects, including the death of normal cells, it is necessary to develop anticancer agents that can target microtubules by specifically acting on cancer cells only. In this study, we identified chemicals that can act as anticancer agents by specifically binding to acetylated microtubules, which are predominant in triple-negative breast cancer (TNBC). The chemical compounds disrupted acetylated microtubule lattices by interfering with microtubule access to alpha-tubulin acetyltransferase 1 (αTAT1), a major acetyltransferase of microtubules, resulting in the increased apoptotic cell death of MDA-MB-231 cells (a TNBC cell line) compared with other cells, such as MCF-10A and MCF-7, which lack microtubule acetylation. Moreover, mouse xenograft experiments showed that treatment with the chemical compounds markedly reduced tumor growth progression. Taken together, the newly identified chemical compounds can be selective for acetylated microtubules and act as potential therapeutic agents against microtubule acetylation enrichment in TNBC.

7.
J Cell Mol Med ; 24(20): 12211-12218, 2020 10.
Article in English | MEDLINE | ID: mdl-32931139

ABSTRACT

Although diesel airborne particulate matter (PM2.5) has been known to play a role in many human diseases, there is no direct evidence that therapeutic drugs or proteins can diminish PM2.5-induced diseases. Nevertheless, studies examining the negative control mechanisms of PM2.5-induced diseases are critical to develop novel therapeutic medications. In this study, the consensus PDZ peptide of ZO-1 inhibited PM2.5-induced inflammatory cell infiltration, pro-inflammatory cytokine gene expression, and TEER in bronchoalveolar lavage (BAL) fluid and AM cells. Our data indicated that the PDZ domain in ZO-1 is critical for regulation of the PM2.5-induced inflammatory microenvironment. Therefore, the PDZ peptide may be a potential therapeutic candidate during PM-induced respiratory diseases.


Subject(s)
Down-Regulation , Gasoline/adverse effects , Particulate Matter/adverse effects , Peptides/pharmacology , Pneumonia/chemically induced , Pneumonia/pathology , Zonula Occludens-1 Protein/chemistry , Amino Acid Motifs , Cell Line , Down-Regulation/drug effects , Humans , PDZ Domains , Particle Size
8.
Cell Mol Life Sci ; 77(20): 4143-4161, 2020 Oct.
Article in English | MEDLINE | ID: mdl-31912196

ABSTRACT

Myofibroblasts are the major cell type that is responsible for increase in the mechanical stiffness in fibrotic tissues. It has well documented that the TGF-ß/Smad axis is required for myofibroblast differentiation under the rigid substrate condition. However, the mechanism driving myofibroblast differentiation in soft substrates remains unknown. In this research, we demonstrated that interaction of yes-associated protein (YAP) and acetylated microtubule via dynein, a microtubule motor protein drives nuclear localization of YAP in the soft matrix, which in turn increased TGF-ß1-induced transcriptional activity of Smad for myofibroblast differentiation. Pharmacological and genetical disruption of dynein impaired the nuclear translocation of YAP and decreased the TGF-ß1-induced Smad activity even though phosphorylation and nuclear localization of Smad occurred normally in α-tubulin acetyltransferase 1 (α-TAT1) knockout cell. Moreover, microtubule acetylation prominently appeared in the fibroblast-like cells nearby the blood vessel in the fibrotic liver induced by CCl4 administration, which was conversely decreased by TGF-ß receptor inhibitor. As a result, quantitative inhibition of microtubule acetylation may be suggested as a new target for overcoming fibrotic diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Dyneins/metabolism , Fibroblasts/metabolism , Microtubules/metabolism , Protein Transport/physiology , Acetylation , Animals , Cell Differentiation/physiology , Cell Line , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Phosphorylation/physiology , Signal Transduction/physiology , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , YAP-Signaling Proteins
9.
J Cell Physiol ; 235(5): 4494-4507, 2020 05.
Article in English | MEDLINE | ID: mdl-31637720

ABSTRACT

Cancer-associated fibroblasts (CAFs) in the tumor microenvironment play major roles in supporting cancer progression. A previous report showed that SPIN90 downregulation is correlated with CAF activation and that SPIN90-deficient CAFs promote breast cancer progression. However, the mechanisms that mediate cancer-stroma interaction and how such interactions regulate cancer progression are not well understood. Here, we show that extra domain A (EDA)-containing fibronectin (FN), FN(+)EDA, produced by mouse embryonic fibroblasts (MEFs) derived from Spin90-knockout (KO) mice increases their own myofibroblast differentiation, which facilitates breast cancer progression. Increased FN(+)EDA in Spin90-KO MEFs promoted fibril formation in the extracellular matrix (ECM) and specifically interacted with integrin α4ß1 as the mediating receptor. Moreover, FN(+)EDA expression by Spin90-KO MEFs increased proliferation, migration, and invasion of breast cancer cells. Irigenin, a specific inhibitor of the interaction between integrin α4ß1 and FN(+)EDA, significantly blocked the effects of FN(+)EDA, such as fibril formation by Spin90-KO MEFs and proliferation, migration, and invasion of breast cancer cells. In orthotopic breast cancer mouse models, irigenin injection remarkably reduced tumor growth and lung metastases. It was supported by that FN(+)EDA in assembled fibrils was accumulated in cancer stroma of human breast cancer patients in which SPIN90 expression was downregulated. Our data suggest that SPIN90 downregulation increases FN(+)EDA and promotes ECM stiffening in breast cancer stroma through an assembly of long FN(+)EDA-rich fibrils; moreover, engagement of the Integrin α4ß1 receptor facilitates breast cancer progression. Inhibitory effects of irigenin on tumor growth and metastasis suggest the potential of this agent as an anticancer therapeutic.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Fibronectins/metabolism , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Female , Fibronectins/genetics , Gene Deletion , Humans , Mammary Neoplasms, Animal , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , Neoplasms, Experimental , Nerve Tissue Proteins/genetics , Up-Regulation
10.
Anim Cells Syst (Seoul) ; 23(6): 414-421, 2019.
Article in English | MEDLINE | ID: mdl-31853379

ABSTRACT

Certain cancer types, including breast cancer, are accompanied with stiffening of the surrounding extracellular matrix (ECM). Previous studies suggest that this stiffened matrix influences cancer cell progression, such as proliferation and invasion, both biochemically and mechanically. However, the contribution of ECM stiffness to cellular response to diverse stresses, which most cancer cells are exposed to, has not been elucidated. In this study, we demonstrate that expression of the Shwachman-Bodian-Diamond syndrome protein (SDBS) in a stiff matrix protects cells from apoptosis induced by environmental stress, including anticancer drugs. Cells cultured on stiff matrices were less apoptotic process induced by serum depletion than those cultured on the soft matrix. Interestingly, knockdown (KD) of SDBS among the apoptosis-related genes significantly increased apoptosis induced by serum depletion in cells cultured in a stiff matrix. Apoptosis of SDBS KD cells in a stiff matrix was significantly inhibited by the caspase 8 inhibitor, indicating that activation of the caspase 8 pathway by SDBS KD is critical for cancer cell apoptosis in stiff matrices. Additionally, we also found that downregulation of SDBS also effectively increased cell death induced by anticancer drugs, including paclitaxel, cisplatin, and eribulin. Taken together, our findings suggest that inhibition of SDBS enhances effective chemotherapy of malignant breast cancer cells in stiff ECM environments.

11.
J Cell Physiol ; 234(6): 9216-9224, 2019 06.
Article in English | MEDLINE | ID: mdl-30341913

ABSTRACT

Alterations in mechanical properties in the extracellular matrix are modulated by myofibroblasts and are required for progressive fibrotic diseases. Recently, we reported that fibroblasts depleted of SPIN90 showed enhanced differentiation into myofibroblasts via increased acetylation of microtubules in the soft matrix; the mechanisms of the underlying signaling network, however, remain unclear. In this study, we determine the effect of depletion of SPIN90 on FAK/ROCK signaling modules. Transcriptome analysis of Spin90 KO mouse embryonic fibroblasts (MEF) and fibroblasts activated by TGF-ß revealed that Postn is the most significantly upregulated gene. Knockdown of Postn by small interfering RNA suppressed cell adhesion and myofibroblastic differentiation and downregulated FAK activity in Spin90 KO MEF. Our results indicate that SPIN90 depletion activates FAK/ROCK signaling, induced by Postn expression, which is critical for myofibroblastic differentiation on soft matrices mimicking the mechanical environment of a normal tissue.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Down-Regulation/genetics , Fibroblasts/metabolism , Focal Adhesion Kinase 1/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , rho-Associated Kinases/metabolism , Animals , Cell Differentiation , Focal Adhesions/metabolism , Mice, Knockout , Myofibroblasts/metabolism
12.
Biochem Biophys Res Commun ; 500(4): 937-943, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29709477

ABSTRACT

Accumulating evidence has shown that matrix stiffening in cancer tissue by the deposition of extracellular matrix (ECM) is closely related with severe tumor progression. However, much less is known about the genes affected by matrix stiffness and its signaling for cancer progression. In the current research, we investigated the differential gene expression of a non-small lung adenocarcinoma cell line, H1299, cultured under the conditions of soft (∼0.5 kPa) and stiff (∼40 kPa) matrices, mimicking the mechanical environments of normal and cancerous tissues, respectively. For integrated transcriptome analysis, the genes identified by ECM stiffening were compared with 8248 genes retrieved from The Cancer Genome Atlas Lung Adenocarcinoma (TCGA). In stiff matrix, 29 genes were significantly upregulated, while 75 genes were downregulated. The screening of hazard ratios for these genes using the Kaplan-Meier Plotter identified 8 genes most closely associated with cancer progression under the condition of matrix stiffening. Among these genes, spindle pole body component 25 homolog (SPC25) was one of the most up-regulated genes in stiff matrix and tumor tissue. Knockdown of SPC25 in H1299 cells using shRNA significantly inhibited cell proliferation with downregulation of the expression of checkpoint protein, Cyclin B1, under the condition of stiff matrix whereas the proliferation rate in soft matrix was not affected by SPC25 silencing. Thus, our findings provide novel key molecules for studying the relationship of extracellular matrix stiffening and cancer progression.


Subject(s)
Cell Proliferation/genetics , Extracellular Matrix/chemistry , Gene Expression Regulation, Neoplastic , Mechanotransduction, Cellular/genetics , Microtubule-Associated Proteins/genetics , Respiratory Mucosa/metabolism , Atlases as Topic , Biomechanical Phenomena , Cell Cycle/genetics , Cell Line, Tumor , Cyclin B1/genetics , Cyclin B1/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression Profiling , HEK293 Cells , Hardness , Humans , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Molecular Sequence Annotation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Respiratory Mucosa/pathology , Transcriptome
13.
Sci Rep ; 7(1): 6847, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28754957

ABSTRACT

Increasing matrix stiffness caused by the extracellular matrix (ECM) deposition surrounding cancer cells is accompanied by epithelial-mesenchymal transition (EMT). Here, we show that expression levels of EMT marker genes along with discoidin domain receptor 2 (DDR2) can increase upon matrix stiffening. DDR2 silencing by short hairpin RNA downregulated EMT markers. Promoter analysis and chromatin immunoprecipitation revealed that c-Myb and LEF1 may be responsible for DDR2 induction during cell culture on a stiff matrix. Mechanistically, c-Myb acetylation by p300, which is upregulated on the stiff matrix, seems to be necessary for the c-Myb-and-LEF1-mediated DDR2 expression. Finally, we found that the c-Myb-DDR2 axis is crucial for lung cancer cell line proliferation and expression of EMT marker genes in a stiff environment. Thus, our results suggest that DDR2 regulation by p300 expression and/or c-Myb acetylation upon matrix stiffening may be necessary for regulation of EMT and invasiveness of lung cancer cells.


Subject(s)
Discoidin Domain Receptor 2/metabolism , Epithelial-Mesenchymal Transition , Extracellular Matrix/chemistry , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Acetylation , Cell Line, Tumor , Cell Proliferation , Discoidin Domain Receptor 2/genetics , HEK293 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Protein Processing, Post-Translational , p300-CBP Transcription Factors/metabolism
14.
Cancer Res ; 77(17): 4710-4722, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28652253

ABSTRACT

Biomechanical remodeling of stroma by cancer-associated fibroblasts (CAF) in early stages of cancer is critical for cancer progression, and mechanical cues such as extracellular matrix stiffness control cell differentiation and malignant progression. However, the mechanism by which CAF activation occurs in low stiffness stroma in early stages of cancer is unclear. Here, we investigated the molecular mechanism underlying CAF regulation by SPIN90 and microtubule acetylation under conditions of mechanically soft matrices corresponding to normal stromal rigidity. SPIN90 was downregulated in breast cancer stroma but not tumor, and this low stromal expression correlated with decreased survival in breast cancer patients. Spin90 deficiency facilitated recruitment of mDia2 and APC complex to microtubules, resulting in increased microtubule acetylation. This increased acetylation promoted nuclear localization of YAP, which upregulated expression of myofibroblast marker genes on soft matrices. Spin90 depletion enhanced tumor progression, and blockade of microtubule acetylation in CAF significantly inhibited tumor growth in mice. Together, our data demonstrate that loss of SPIN90-mediated microtubule acetylation is a key step in CAF activation in low stiffness stroma. Moreover, correlation among these factors in human breast cancer tissue supports the clinical relevance of SPIN90 and microtubule acetylation in tumor development. Cancer Res; 77(17); 4710-22. ©2017 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Breast Neoplasms/pathology , Fibroblasts/pathology , Microtubules/pathology , Muscle Proteins/metabolism , Nerve Tissue Proteins/physiology , Stromal Cells/pathology , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Differentiation , Disease Progression , Female , Fibroblasts/metabolism , Humans , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Muscle Proteins/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphoproteins/metabolism , Stromal Cells/metabolism , Transcription Factors , Tumor Cells, Cultured , Tumor Microenvironment , YAP-Signaling Proteins
15.
Biochem Biophys Res Commun ; 482(1): 8-14, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27836544

ABSTRACT

Microtubules are required for diverse cellular processes, and abnormal regulation of microtubule dynamics is closely associated with severe diseases including malignant tumors. In this study, we report that α-tubulin N-acetyltransferase (αTAT1), a regulator of α-tubulin acetylation, is required for colon cancer proliferation and invasion via regulation of Wnt1 and its downstream genes expression. Public transcriptome analysis showed that expression of ATAT1 is specifically upregulated in colon cancer tissue. A knockout (KO) of ATAT1 in the HCT116 colon cancer cell line, using the CRISPR/Cas9 system showed profound inhibition of proliferative and invasive activities of these cancer cells. Overexpression of αTAT1 or the acetyl-mimic K40Q α-tubulin mutant in αTAT1 KO cells restored the invasiveness, indicating that microtubule acetylation induced by αTAT1 is critical for HCT116 cell invasion. Analysis of colon cancer-related gene expression in αTAT1 KO cells revealed that the loss of αTAT1 decreased the expression of WNT1. Mechanistically, abrogation of tubulin acetylation by αTAT1 knockout inhibited localization of ß-catenin to the plasma membrane and nucleus, thereby resulting in the downregulation of Wnt1 and of its downstream genes including CCND1, MMP-2, and MMP-9. These results suggest that αTAT1-mediated Wnt1 expression via microtubule acetylation is important for colon cancer progression.


Subject(s)
Acetyltransferases/genetics , Cell Proliferation/genetics , Colonic Neoplasms/genetics , Microtubules/genetics , Wnt Signaling Pathway/genetics , Wnt1 Protein/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Down-Regulation/genetics , Gene Knockout Techniques , Humans , Neoplasm Invasiveness
16.
Sci Rep ; 6: 27054, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27270970

ABSTRACT

Respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), and lung infections have critical consequences on mortality and morbidity in humans. The aims of the present study were to examine the mechanisms by which CXCL12 affects MUC1 transcription and airway inflammation, which depend on activator of G-protein signaling (AGS) 3 and to identify specific molecules that suppress CXCL12-induced airway inflammation by acting on G-protein-coupled receptors. Herein, AGS3 suppresses CXCL12-mediated upregulation of MUC1 and TNFα by regulating Gαi. We found that the G-protein regulatory (GPR) motif peptide in AGS3 binds to Gαi and downregulates MUC1 expression; in contrast, this motif upregulates TNFα expression. Mutated GPR Q34A peptide increased the expression of MUC1 and TGFß but decreased the expression of TNFα and IL-6. Moreover, CXCR4-induced dendritic extensions in 2D and 3D matrix cultures were inhibited by the GPR Q34A peptide compared with a wild-type GPR peptide. The GPR Q34A peptide also inhibited CXCL12-induced morphological changes and inflammatory cell infiltration in the mouse lung, and production of inflammatory cytokines in bronchoalveolar lavage (BAL) fluid and the lungs. Our data indicate that the GPR motif of AGS3 is critical for regulating MUC1/Muc1 expression and cytokine production in the inflammatory microenvironment.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/physiology , Respiratory Mucosa/metabolism , Transcriptional Activation , Actins/metabolism , Amino Acid Sequence , Animals , Cell Line , Chemokine CXCL12/physiology , Gene Expression , Humans , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/immunology , Lung/metabolism , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Mice, Inbred C57BL , Mucin-1/genetics , Mucin-1/metabolism , Peptide Fragments/physiology , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Receptors, CXCR4/metabolism , Respiratory Mucosa/immunology , Up-Regulation
17.
Sci Rep ; 6: 21564, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26877098

ABSTRACT

Dynamic interaction between cancer cells and the surrounding microenvironment is critical for cancer progression via changes in cellular behavior including alteration of secreted molecules. However, the molecular mechanisms underlying the influence exerted by the cancer microenvironment on secretion of molecules during cancer progression remain largely unknown. In this study, we report that secretion of spingsine-1-phosphate (S1P) and its regulator, SphK1 expression is dependent of the substrate rigidity, which is critical for the balance between cancer cell invasion and adhesion. Conditioned media (CM) of MDA-MB-231, an aggressive breast cancer cell obtained from soft substrate (~0.5 kPa) induced chemo-attractive invasion, while CM obtained from stiff substrate (~2.5 kPa) increased cell adhesion instead. We found that the expression of SphK1 is upregulated in the stiff substrate, resulting in an increase in S1P levels in the CM. We also found that upregulation of SphK1 expression in the stiff substrate is dominant in metastatic cancer cells but not in primary cancer cells. These results suggest that alterations in the mechanical environment of the ECM surrounding the tumor cells actively regulate cellular properties such as secretion, which in turn, may contribute to cancer progression.


Subject(s)
Breast Neoplasms/physiopathology , Cell Adhesion , Extracellular Matrix/physiology , Lysophospholipids/metabolism , Neoplasm Metastasis/physiopathology , Sphingosine/analogs & derivatives , Biomechanical Phenomena , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Female , Humans , Neoplasm Invasiveness/physiopathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/metabolism , Tumor Microenvironment
18.
Oncotarget ; 7(14): 17829-43, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26893363

ABSTRACT

Mammalian diaphanous-related formin 1 (mDia1) expression has been linked with progression of malignant cancers in various tissues. However, the precise molecular mechanism underlying mDia1-mediated invasion in cancer cells has not been fully elucidated. In this study, we found that mDia1 is upregulated in invasive breast cancer cells. Knockdown of mDia1 in invasive breast cancer profoundly reduced invasive activity by controlling cellular localization of membrane type 1-matrix metalloproteinase (MT1-MMP) through interaction with microtubule tracks. Gene silencing and ectopic expression of the active form of mDia1 showed that mDia1 plays a key role in the intracellular trafficking of MT1-MMP to the plasma membrane through microtubules. We also demonstrated that highly invasive breast cancer cells possessed invasive activity in a 3D culture system, which was significantly reduced upon silencing mDia1 or MT1-MMP. Furthermore, mDia1-deficient cells cultured in 3D matrix showed impaired expression of the cancer stem cell marker genes, CD44 and CD133. Collectively, our findings suggest that regulation of cellular trafficking and microtubule-mediated localization of MT1-MMP by mDia1 is likely important in breast cancer invasion through the expression of cancer stem cell genes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Matrix Metalloproteinase 14/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Female , Formins , Humans , MCF-7 Cells , Matrix Metalloproteinase 14/genetics , Microtubules/metabolism , Neoplasm Invasiveness , Transfection
19.
Int J Cancer ; 135(11): 2547-57, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24740739

ABSTRACT

Discoidin domain receptors (DDRs) are unusual receptor tyrosine kinases (RTKs) that are activated by fibrillar collagens instead of soluble growth factors. DDRs play an important role in various cellular functions and disease processes, including malignant progression. Compared to other RTKs, DDRs have relatively long juxtamembrane domains, which are believed to contribute to receptor function. Despite this possibility, the function and mechanism of the juxtamembrane domain of DDRs have not yet been fully elucidated. In this study, we found that the cytoplasmic juxtamembrane 2 (JM2) region of DDR2 contributed to receptor dimerization, which is critical for receptor activation in response to collagen stimulation. A collagen-binding assay showed that JM2 was required for efficient binding of collagen to the discoidin (DS) domain. Immunohistochemical analysis of DDR2 expression using a tissue microarray demonstrated that DDR2 was overexpressed in several carcinoma tissues, including bladder, testis, lung, kidney, prostate and stomach. In H1299 cells, inhibition of DDR2 activity by overexpressing the juxtamembrane domain containing JM2 suppressed collagen-induced colony formation, cell proliferation and invasion via the inhibition of matrix metalloproteinase-2 and matrix metalloproteinase-9. Taken together, our results suggest that JM2-mediated dimerization is likely to be essential for DDR2 activation and cancer progression. Thus, inhibition of DDR2 function using a JM2-containing peptide might be a useful strategy for the treatment of DDR2-positive cancers.


Subject(s)
Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Collagen/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Binding Sites , Blotting, Western , Cell Adhesion , Cross-Linking Reagents/pharmacology , Discoidin Domain Receptors , Disease Progression , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Microscopy, Fluorescence , Phosphorylation , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Signal Transduction , Tumor Cells, Cultured
20.
Int J Mol Med ; 31(5): 1113-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23546533

ABSTRACT

The collagen matrix constitutes the primary extracellular matrix (ECM) portion of mammalian connective tissues in which the interaction of the cell and the surrounding collagen fibers has a significant impact on cell and tissue physiology, including morphogenesis, development and motility. Discoidin domain receptors (DDR1 and DDR2) have been identified as the receptor tyrosine kinases that are activated upon collagen binding. However, there is a lack of evidence regarding the effect of DDRs on the mechanical interaction between fibroblasts and ECM. In this study, we demonstrated that one of the major phosphotyrosine proteins in human fibroblasts during 3D collagen matrix polymerization is DDR2. Treatment of fibroblasts in 3D collagen matrices with platelet-derived growth factor (PDFG) has been shown to increase DDR2 phosphorylation. Silencing of DDR2 with siRNA in fibroblasts significantly reduced the number of dendritic extensions regardless of whether cells were cultured in the collagen or fibronectin 3D matrices. Decreasing dendritic extensions in DDR2-silenced cells has also been shown to decrease the ability of fibroblast entanglement to collagen fibrils in 3D collagen matrices. Finally, we also showed that the silencing of DDR2 decreased the cell migration in 3D nested collagen matrices but had no effect on 3D floating matrix contraction. Collectively, these results suggest that DDR2 functioning is required for the membrane dynamics to control the mechanical attachment of fibroblasts to the 3D collagen matrices in an integrin-independent manner.


Subject(s)
Collagen/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell-Matrix Junctions/drug effects , Cell-Matrix Junctions/metabolism , Dendrites/drug effects , Dendrites/metabolism , Discoidin Domain Receptors , Fibroblasts/drug effects , Gene Silencing/drug effects , Humans , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...