Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Theranostics ; 11(16): 8092-8111, 2021.
Article in English | MEDLINE | ID: mdl-34335982

ABSTRACT

Active c-Src non-receptor tyrosine kinase localizes to the plasma membrane via N-terminal lipid modification. Membranous c-Src causes cancer initiation and progression. Even though transmembrane 4 L six family member 5 (TM4SF5), a tetraspan(in), can be involved in this mechanism, the molecular and structural influence of TM4SF5 on c-Src remains unknown. Methods: Here, we investigated molecular and structural details by which TM4SF5 regulated c-Src devoid of its N-terminus and how cell-penetrating peptides were able to interrupt c-Src activation via interference of c-Src-TM4SF5 interaction in hepatocellular carcinoma models. Results: The TM4SF5 C-terminus efficiently bound the c-Src SH1 kinase domain, efficiently to the inactively-closed form. The complex involved protein tyrosine phosphatase 1B able to dephosphorylate Tyr530. The c-Src SH1 domain alone, even in a closed form, bound TM4SF5 to cause c-Src Tyr419 and FAK Y861 phosphorylation. Homology modeling and molecular dynamics simulation studies predicted the directly interfacing residues, which were further validated by mutational studies. Cell penetration of TM4SF5 C-terminal peptides blocked the interaction of TM4SF5 with c-Src and prevented c-Src-dependent tumor initiation and progression in vivo. Conclusions: Collectively, these data demonstrate that binding of the TM4SF5 C-terminus to the kinase domain of inactive c-Src leads to its activation. Because this binding can be abolished by cell-penetrating peptides containing the TM4SF5 C-terminus, targeting this direct interaction may be an effective strategy for developing therapeutics that block the development and progression of hepatocellular carcinoma.


Subject(s)
CSK Tyrosine-Protein Kinase/metabolism , Carcinoma, Hepatocellular/metabolism , Membrane Proteins/metabolism , CSK Tyrosine-Protein Kinase/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/physiology , Genes, src/genetics , Genes, src/physiology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/physiology , Peptides/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tetraspanins/genetics , Tetraspanins/metabolism
2.
Oncotarget ; 8(8): 13277-13292, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28129652

ABSTRACT

The transmembrane 4 L six family proteins TM4SF1, TM4SF4, and TM4SF5 share 40-50% overall sequence identity, but their C-terminus identity is limited. It may be likely that the C-termini of the members are important and unique for own regulatory functions. We thus examined how the TM4SF5 C-terminus affected cellular functions differentially from other family members. Using colon cancer cells expressing wildtype (WT), C-terminus-deleted, or chimeric mutants, diverse cellular functions were explored in 2-dimensional (2D) and 3-dimensional (3D) condition. The C-termini of the proteins were relatively comparable with respect to 2D cell proliferation, although each C-terminal-deletion mutant exhibited increased proliferation relative to the WT. Using chimeric constructs, we found that the TM4SF5 C-terminus was critical for regulating the diverse metastatic functions of TM4SF5, and could positively replace the C-termini of other family members. Replacement of the TM4SF1 or TM4SF4 C-terminus with that of TM4SF5 increased spheroids growth, transwell migration, and invasive dissemination from spheroids in 3D collagen gels. TM4SF5-mediated effects required its extracellular loop 2 linked to the C-terminus via the transmembrane domain 4, with causing c-Src activation. Altogether, the C-terminus of TM4SF5 appears to mediate pro-migratory roles, depending on a structural relay from the second extracellular loop to the C-terminus.


Subject(s)
Antigens, Surface/genetics , Cell Proliferation/genetics , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mutation , Neoplasm Proteins/genetics , Animals , Antigens, Surface/metabolism , Blotting, Western , Cell Culture Techniques , Cell Line, Tumor , Cell Movement/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , HT29 Cells , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Neoplasm Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/metabolism , Transplantation, Heterologous
4.
Oncotarget ; 6(25): 21655-74, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26091349

ABSTRACT

The adhesion properties of cells are involved in tumor metastasis. Although KRS at the plasma membrane is shown important for cancer metastasis, additionally to canonical roles of cytosolic KRS in protein translation, how KRS and its downstream effectors promote the metastatic migration remains unexplored. Disseminative behaviors (an earlier metastatic process) of colon cancer cell spheroids embedded in 3D collagen gels were studied with regards to cell adhesion properties, and relevance in KRS(-/+) knocked-down animal and clinical colon cancer tissues. Time-lapse imaging revealed KRS-dependent cell dissemination from the spheroids, whereas KRS-suppressed spheroids remained static due to the absence of outbound movements supported by cell-extracellular matrix (ECM) adhesion. While keeping E-cadherin at the outward disseminative cells, KRS caused integrin-involved intracellular signaling for ERK/c-Jun, paxillin, and cell-ECM adhesion-mediated signaling to modulate traction force for crawling movement. KRS-suppressed spheroids became disseminative following ERK or paxillin re-expression. The KRS-dependent intracellular signaling activities correlated with the invasiveness in clinical colon tumor tissues and in KRS(-/+) knocked-down mice tissues. Collectively, these observations indicate that KRS at the plasma membrane plays new roles in metastatic migration as a signaling inducer, and causes intracellular signaling for cancer dissemination, involving cell-cell and cell-ECM adhesion, during KRS-mediated metastasis.


Subject(s)
Collagen Type I/metabolism , Colonic Neoplasms/enzymology , Lysine-tRNA Ligase/metabolism , Animals , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Colonic Neoplasms/pathology , Cytosol/metabolism , Extracellular Matrix/metabolism , Female , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique, Indirect , HCT116 Cells , Humans , Mice , Neoplasm Metastasis , Paxillin/metabolism , Phosphorylation , Protein Biosynthesis , Signal Transduction
5.
Hepatology ; 61(6): 1978-97, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25627085

ABSTRACT

UNLABELLED: Tumor metastasis involves circulating and tumor-initiating capacities of metastatic cancer cells. Epithelial-mesenchymal transition (EMT) is related to self-renewal capacity and circulating tumor cell (CTC) characteristics for tumor metastasis. Although tumor metastasis is a life-threatening, complicated process that occurs through circulation of tumor cells, mechanistic aspects of self-renewal and circulating capacities have been largely unknown. Hepatic transmembrane 4 L six family member 5 (TM4SF5) promotes EMT for malignant growth and migration, so it was rationalized that TM4SF5, as a hepatocellular carcinoma (HCC) biomarker, might be important for metastatic potential. Here, self-renewal capacity by TM4SF5 was mechanistically explored using hepatocarcinoma cells with or without TM4SF5 expression, and we explored whether they became CTCs using mouse liver-orthotopic model systems. We found that TM4SF5-dependent sphere growth correlated with CD24(-) , aldehyde dehydrogenase (ALDH) activity, as well as a physical association between CD44 and TM4SF5. Interaction between TM4SF5 and CD44 was through their extracellular domains with N-glycosylation modifications. TM4SF5/CD44 interaction activated proto-oncogene tyrosine-protein kinase Src (c-Src)/signal transducer and activator of transcription 3 (STAT3)/Twist-related protein 1 (Twist1)/B-cell-specific Moloney murine leukemia virus integration site 1 (Bmi1) signaling for spheroid formation, whereas disturbing the interaction, expression, or activity of any component in this signaling pathway inhibited spheroid formation. In serial xenografts using 200∼5,000 cells per injection, TM4SF5-positive tumors exhibited subpopulations with locally increased CD44 expressions, supporting for tumor cell differentiation. TM4SF5-positive, but not TM4SF5- or CD44-knocked-down, cells were identified circulating in blood 4-6 weeks after orthotopic liver injection using in vivo laser scanning endomicroscopy. Anti-TM4SF5 reagent blocked their metastasis to distal intestinal organs. CONCLUSION: TM4SF5 promotes self-renewal and CTC properties supported by TM4SF5(+) /CD44(+(TM4SF5-bound)) /ALDH(+) /CD24(-) markers during HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hyaluronan Receptors/metabolism , Liver Neoplasms, Experimental/metabolism , Membrane Proteins/metabolism , Neoplastic Cells, Circulating/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Spheroids, Cellular , Twist-Related Protein 1/metabolism , src-Family Kinases/metabolism
6.
PLoS One ; 9(7): e102817, 2014.
Article in English | MEDLINE | ID: mdl-25033048

ABSTRACT

Transmembrane 4 L6 family member 5 (TM4SF5) is overexpressed during CCl4-mediated murine liver fibrosis and in human hepatocellular carcinomas. The tetraspanins form tetraspanin-enriched microdomains (TEMs) consisting of large membrane protein complexes on the cell surface. Thus, TM4SF5 may be involved in the signal coordination that controls liver malignancy. We investigated the relationship between TM4SF5-positive TEMs with liver fibrosis and tumorigenesis, using normal Chang hepatocytes that lack TM4SF5 expression and chronically TGFß1-treated Chang cells that express TM4SF5. TM4SF5 expression is positively correlated with tumorigenic CD151 expression, but is negatively correlated with tumor-suppressive CD63 expression in mouse fibrotic and human hepatic carcinoma tissues, indicating cooperative roles of the tetraspanins in liver malignancies. Although CD151 did not control the expression of TM4SF5, TM4SF5 appeared to control the expression levels of CD151 and CD63. TM4SF5 interacted with CD151, and caused the internalization of CD63 from the cell surface into late lysosomal membranes, presumably leading to terminating the tumor-suppressive functions of CD63. TM4SF5 could overcome the tumorigenic effects of CD151, especially cell migration and extracellular matrix (ECM)-degradation. Taken together, TM4SF5 appears to play a role in liver malignancy by controlling the levels of tetraspanins on the cell surface, and could provide a promising therapeutic target for the treatment of liver malignancies.


Subject(s)
Cell Movement/physiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Membrane Proteins/metabolism , Neoplasm Invasiveness/pathology , Tetraspanin 24/metabolism , Tetraspanin 30/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Phenotype , Tetraspanins/metabolism , Transforming Growth Factor beta1/metabolism
7.
Biochem J ; 462(1): 89-101, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24897542

ABSTRACT

TM4SF5 (transmembrane 4 L six family member 5) is involved in EMT (epithelial-mesenchymal transition) for liver fibrosis and cancer metastasis; however, the function(s) of TM4SF5 during embryogenesis remains unknown. In the present study the effects of TM4SF5 on embryogenesis of zebrafish were investigated. tm4sf5 mRNA was expressed in the posterior somites during somitogenesis and in whole myotome 1 dpf (day post-fertilization). tm4sf5 suppression impaired development of the trunk with aberrant morphology of muscle fibres and altered expression of integrin α5. The arrangement and adhesion of muscle cells were abnormally disorganized in tm4sf5 morphants with reduced muscle fibre masses, where integrin α5-related signalling molecules, including fibronectin, FAK (focal adhesion kinase), vinculin and actin were aberrantly localized, compared with those in control fish. Aberrant muscle developments in tm4sf5 morphants were recovered by additional tm4sf5 or integrin α5 mRNA injection. Such a role for TM4SF5 was observed in the differentiation of C2C12 mouse myoblast cells to multinuclear muscle cells. Taken together, the results show that TM4SF5 controls muscle differentiation via co-operation with integrin α5-related signalling.


Subject(s)
Integrin alpha5/physiology , Membrane Proteins/genetics , Muscle Development/physiology , Animals , Cell Differentiation , Cell Movement , Cells, Cultured , Epithelial-Mesenchymal Transition , Integrin alpha5/biosynthesis , Membrane Proteins/biosynthesis , Mice , Signal Transduction/physiology , Somites/metabolism , Zebrafish/embryology
8.
Mol Cell Biol ; 34(16): 2946-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24912675

ABSTRACT

TM4SF5 overexpressed in hepatocellular carcinoma activates focal adhesion kinase (FAK) during tumor cell migration. However, it remains unknown how TM4SF5 in hepatocellular carcinoma cells compromises with immune actions initiated by extracellular cytokines. Normal and cancerous hepatocytes with or without TM4SF5 expression were analyzed for the effects of cytokine signaling activity on TM4SF5/FAK signaling and metastatic potential. We found that interleukin-6 (IL-6) was differentially expressed in hepatocytes depending on cancerous malignancy and TM4SF5 expression. IL-6 treatment activated FAK and STAT3 and enhanced focal adhesion (FA) formation in TM4SF5-null cells, but it decreased TM4SF5-dependent FAK activity and FA formation in SNU761-TM4SF5 cells. STAT3 suppression abolished the IL-6-mediated effects in normal Chang cells, but it did not recover the TM4SF5-dependent FAK activity that was inhibited by IL-6 treatment in cancerous SNU761-TM4SF5 cells. In addition, modulation of FAK activity did not change the IL-6-mediated STAT3 activity in either the Chang or SNU761 cell system. TM4SF5 expression in SNU761 cells caused invasive extracellular matrix degradation negatively depending on IL-6/IL-6 receptor (IL-6R) signaling. Thus, it is likely that hepatic cancer cells adopt TM4SF5-dependent FAK activation and metastatic potential by lowering IL-6 expression and avoiding its immunological action through the IL-6-STAT3 pathway.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/genetics , Interleukin-6/metabolism , Liver Neoplasms/immunology , Membrane Proteins/genetics , STAT3 Transcription Factor/metabolism , Tumor Escape , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/immunology , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesions/genetics , Hepatocytes/cytology , Humans , Interleukin-6/biosynthesis , Interleukin-6/pharmacology , Liver Neoplasms/genetics , Membrane Proteins/biosynthesis , Neoplasm Metastasis , Phosphorylation , Receptors, Interleukin-6/immunology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Escape/genetics , Tumor Escape/immunology
9.
Biochim Biophys Acta ; 1843(9): 2037-54, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24861866

ABSTRACT

Although an in vitro 3D environment cannot completely mimic the in vivo tumor site, embedding tumor cells in a 3D extracellular matrix (ECM) allows for the study of cancer cell behaviors and the screening of anti-metastatic reagents with a more in vivo-like context. Here we explored the behaviors of MDA-MB-231 breast cancer cells embedded in 3D collagen I. Diverse tumor environmental conditions (including cell density, extracellular acidity, or hypoxia as mimics for a continuous tumor growth) reduced JNKs, enhanced TGFß1/Smad signaling activity, induced Snail1, and reduced cortactin expression. The reduced JNKs activity blocked efficient formation of invadopodia labeled with actin, cortactin, or MT1-MMP. JNKs inactivation activated Smad2 and Smad4, which were required for Snail1 expression. Snail1 then repressed cortactin expression, causing reduced invadopodia formation and prominent localization of MT1-MMP at perinuclear regions. MDA-MB-231 cells thus exhibited less efficient collagen I degradation and invasion in 3D collagen I upon JNKs inhibition. These observations support a signaling network among JNKs, Smads, Snail1, and cortactin to regulate the invasion of MDA-MB-231 cells embedded in 3D collagen I, which may be targeted during screening of anti-invasion reagents.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Collagen Type I/pharmacology , Cortactin/metabolism , Pseudopodia/metabolism , Transcription Factors/metabolism , Tumor Microenvironment/drug effects , Actins/metabolism , Animals , Breast Neoplasms/enzymology , Cattle , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Cell Nucleus/metabolism , Cortactin/genetics , Female , Gels , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 14/metabolism , Neoplasm Invasiveness , Phosphoserine/metabolism , Protein Transport , Proto-Oncogene Proteins c-jun/metabolism , Pseudopodia/drug effects , Signal Transduction , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription, Genetic , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL