Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 8675, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33883672

ABSTRACT

In epithelial cancers, the epidermal growth factor receptor (EGFR) and integrin α6ß4 are frequently overexpressed and found to synergistically activate intracellular signaling pathways that promote cell proliferation and migration. In cancer cells, the ß4 subunit is phosphorylated at tyrosine residues not normally recognized as kinase substrates; however, the function of these phosphotyrosine residues in cancer cells is a subject of much debate. In EGFR-overexpressing carcinoma cells, we found that the Src family kinase (SFK) inhibitor PP2 reduces ß4 tyrosine phosphorylation following the activation of EGFR. However, siRNA mediated knockdown of the SFKs Src, Fyn, Yes and Lyn, individually or in combination, did not affect the EGF-induced phosphorylation of ß4. Using phospho-peptide affinity chromatography and mass spectrometry, we found that PLCγ1 binds ß4 at the phosphorylated residues Y1422/Y1440, but were unable to verify this interaction in A431 carcinoma cells that overexpress the EGFR. Furthermore, using A431 cells devoid of ß4 or reconstituted with phenylalanine specific mutants of ß4, the activation of several downstream signaling pathways, including PLCγ/PKC, MAPK and PI3K/Akt, were not substantially affected. We conclude that tyrosine-phosphorylated ß4 does not enhance EGFR-mediated signaling in EGFR-overexpressing cells, despite the fact that this integrin subunit is highly tyrosine phosphorylated in these cells.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Integrin beta4/metabolism , Skin Neoplasms/metabolism , Tyrosine/metabolism , Animals , Cell Line, Tumor , Humans , Integrin beta4/physiology , Mass Spectrometry , Phosphorylation , Phosphotyrosine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Skin Neoplasms/genetics
2.
Development ; 147(4)2020 02 17.
Article in English | MEDLINE | ID: mdl-31988184

ABSTRACT

Integrin dimers α3/ß1, α6/ß1 and α6/ß4 are the mammary epithelial cell receptors for laminins, which are major components of the mammary basement membrane. The roles of specific basement membrane components and their integrin receptors in the regulation of functional gland development have not been analyzed in detail. To investigate the functions of laminin-binding integrins, we obtained mutant mice with mammary luminal cell-specific deficiencies of the α3 and α6 integrin chains generated using the Cre-Lox approach. During pregnancy, mutant mice displayed decreased luminal progenitor activity and retarded lobulo-alveolar development. Mammary glands appeared functional at the onset of lactation in mutant mice; however, myoepithelial cell morphology was markedly altered, suggesting cellular compensation mechanisms involving cytoskeleton reorganization. Notably, lactation was not sustained in mutant females, and the glands underwent precocious involution. Inactivation of the p53 gene rescued the growth defects but did not restore lactogenesis in mutant mice. These results suggest that the p53 pathway is involved in the control of mammary cell proliferation and survival downstream of laminin-binding integrins, and underline an essential role of cell interactions with laminin for lactogenic differentiation.


Subject(s)
Integrins/physiology , Lactation , Mammary Glands, Animal/physiology , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Survival , Cytoskeleton/physiology , Disease Progression , Female , Gene Deletion , Hormones/physiology , Integrin alpha3/physiology , Integrin alpha6/physiology , Integrin beta1/physiology , Integrin beta4/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Mutant Strains , Mutation , Neoplastic Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Ovary/physiology , Phenotype , Pregnancy , Pregnancy, Animal , Prognosis , Protein Binding , Protein Multimerization
3.
Sheng Li Xue Bao ; 70(5): 504-510, 2018 Oct 25.
Article in Chinese | MEDLINE | ID: mdl-30377689

ABSTRACT

Integrin is a transmembrane receptor that mediates the connection between cells and their external environment, such as extracellular matrix (ECM). Integrin ß4 (ITGß4) plays a number of functions due to its special structures: forms α6ß4 with ITGα6 subunit and participates in the formation of hemidesmosomes; mediates cell-to-cell matrix interaction and cell-to-cell interaction, cell proliferation and survival, as well as migration and invasion. Also, ITGß4 participates in various disease processes by activating multiple signaling pathways. In this paper, the structure, physiological function and function of ITGß4 in respiratory system, tumor, nervous system and other related diseases will be reviewed.


Subject(s)
Integrin beta4/physiology , Cell Communication , Cell Movement , Cell Proliferation , Hemidesmosomes , Humans , Integrin alpha6beta4 , Signal Transduction
4.
J Leukoc Biol ; 103(5): 897-908, 2018 05.
Article in English | MEDLINE | ID: mdl-29393977

ABSTRACT

Airway epithelial cells play a significant role in the pathogenesis of asthma. Although the structural and functional defects of airway epithelial cells have been postulated to increase asthma susceptibility and exacerbate asthma severity, the mechanism and implication of these defects remain uncertain. Integrin ß4 (ITGB4) is a structural adhesion molecule that is downregulated in the airway epithelium of asthma patients. In this study, we demonstrated that ITGB4 deficiency leads to severe allergy-induced airway inflammation and airway hyper-responsiveness (AHR) in mice. After house dust mite (HDM) challenge, epithelial cell-specific ITGB4-deleted mice showed increased lymphocyte, eosinophil, and neutrophil infiltration into lung compared with that of the wild-type mice. ITGB4 deficiency also resulted in increased expression of the Th2 cytokine IL-4, IL-13, and the Th17 cytokine IL-17A in the lung tissue and in the T cells after HDM challenge. The aggravated inflammation in ITGB4 defect mice was partly caused by enhanced disrupted epithelial barrier integrity after HDM stress, which induced the increased thymic stromal lymphopoietin secretion from airway epithelial cells. This study therefore demonstrates that ITGB4 plays a pivotal role in containing allergen-mediated lung inflammation and airway hyper-responsiveness in allergic asthma.


Subject(s)
Allergens/adverse effects , Integrin beta4/physiology , Pneumonia/etiology , Pyroglyphidae/pathogenicity , Respiratory Hypersensitivity/etiology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/pathology , Respiratory Hypersensitivity/pathology
5.
Oncogene ; 35(31): 4112-21, 2016 08 04.
Article in English | MEDLINE | ID: mdl-26751774

ABSTRACT

MUC5AC is a secretory mucin aberrantly expressed in various cancers. In lung cancer, MUC5AC is overexpressed in both primary and metastatic lesions; however, its functional role is not well understood. The present study was aimed at evaluating mechanistic role of MUC5AC on metastasis of lung cancer cells. Clinically, the overexpression of MUC5AC was observed in lung cancer patient tissues and was associated with poor survival. In addition, the overexpression of Muc5ac was also observed in genetically engineered mouse lung adenocarcinoma tissues (Kras(G12D); Trp53(R172H/+); AdCre) in comparison with normal lung tissues. Our functional studies showed that MUC5AC knockdown resulted in significantly decreased migration in two lung cancer cell lines (A549 and H1437) as compared with scramble cells. Expression of integrins (α5, ß1, ß3, ß4 and ß5) was decreased in MUC5AC knockdown cells. As both integrins and MUC5AC have a von Willebrand factor domain, we assessed for possible interaction of MUC5AC and integrins in lung cancer cells. MUC5AC strongly interacted only with integrin ß4. The co-localization of MUC5AC and integrin ß4 was observed both in A549 lung cancer cells as well as genetically engineered mouse adenocarcinoma tissues. Activated integrins recruit focal adhesion kinase (FAK) that mediates metastatic downstream signaling pathways. Phosphorylation of FAK (Y397) was decreased in MUC5AC knockdown cells. MUC5AC/integrin ß4/FAK-mediated lung cancer cell migration was confirmed through experiments utilizing a phosphorylation (Y397)-specific FAK inhibitor. In conclusion, overexpression of MUC5AC is a poor prognostic marker in lung cancer. MUC5AC interacts with integrin ß4 that mediates phosphorylation of FAK at Y397 leading to lung cancer cell migration.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/physiology , Integrin beta4/physiology , Lung Neoplasms/pathology , Mucin 5AC/physiology , Signal Transduction/physiology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Humans , Integrin beta4/analysis , Male , Mice , Mucin 5AC/analysis , Phosphorylation
6.
Oncogene ; 34(34): 4491-9, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-25531330

ABSTRACT

Epstein-Barr virus (EBV)-encoded Latent Membrane Protein 2A (LMP2A) is an EBV latency-associated protein regularly expressed in nasopharyngeal carcinoma (NPC). In B cells, LMP2A activity resembles that of a constitutively activated antigen receptor, which recruits the Syk tyrosine kinase to activate a set of downstream signaling pathways. LMP2A also downregulates cellular Syk levels. In the present study, we demonstrate that Syk interacts with the integrin ß4 subunit (ITGß4) of integrin α6ß4 in epithelial cells and that concurrent LMP2A expression interferes with this interaction by competitive binding to Syk. We find that both Syk and LMP2A have an effect on ITGß4 cell surface expression. However, in LMP2A expressing cells, ITGß4 remains concentrated at the cellular protrusions, an expression pattern characteristic of motile cells, including NPC-derived epithelial cells. This effect of LMP2A on ITGß4 localization is associated with a greater propensity for migration and invasion in-vitro, and may contribute to the invasive property of LMP2A-expressing NPC.


Subject(s)
Cell Movement , Integrin beta4/physiology , Intracellular Signaling Peptides and Proteins/physiology , Nasopharyngeal Neoplasms/pathology , Protein-Tyrosine Kinases/physiology , Viral Matrix Proteins/physiology , Amino Acid Sequence , Cell Line, Tumor , Humans , Molecular Sequence Data , Neoplasm Invasiveness , Syk Kinase
7.
Breast Cancer Res ; 16(5): 459, 2014.
Article in English | MEDLINE | ID: mdl-25606594

ABSTRACT

Following a highly dynamic and complex dialogue between the epithelium and the surrounding microenvironment, the mammary gland develops into a branching structure during puberty, buds during pregnancy, forms intricate polar acini during lactation and, once the babies are weaned, remodels and involutes. At every stage of menstrual and pregnancy cycles, interactions between the cells and the extracellular matrix (ECM) and homotypic and heterotypic cell­cell interactions give rise to the architecture and function of the gland at that junction. These orchestrated programs would not be possible without the important role of the ECM receptors, integrins being the prime examples. The ECM­integrin axis regulates many crucial cellular functions including survival, migration and quiescence; the imbalance in any of these processes could contribute to oncogenesis. In this review we spotlight the involvement of two prominent integrin subunits, ß1 and ß4 integrins, in cross-talk with tyrosine kinase receptors, and we discuss the roles of these integrin subunits in the biology of normal breast differentiation and as potential prognostic and therapeutic targets in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Integrin beta1/physiology , Integrin beta4/physiology , Mammary Glands, Human/growth & development , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Adhesion , Disease Progression , Drug Resistance, Neoplasm , Female , Humans , Mammary Glands, Human/metabolism
8.
J Dermatol Sci ; 61(3): 169-79, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21282044

ABSTRACT

BACKGROUND: Adhesion of keratinocytes is crucial for maintaining the integrity of the skin, as demonstrated by the number of dermatological disorders of genetic origin that are associated with a defect of basal keratinocyte adhesion. Integrins are the main component of the molecular networks involved in this phenomenon, but there are many others. In a recent description of proteins associated to caveolae at the plasma membrane of human basal epidermal cells, we demonstrated that CD98hc is localized with ß1 integrin. OBJECTIVES: We investigated the CD98hc proteins interactions and the role of CD98hc in keratinocyte adhesion. METHODS: CD98hc protein interaction was identified following co-immunoprecipitation and proteomic analysis using LTQ-FT mass spectrometer. Extinction of CD98hc gene expression using specific short hairpin RNA or over-expression of CD98hc lacking the ß1 integrin binding site was used to evaluate the role of this protein in keratinocyte fate. RESULTS: We show that CD98hc forms molecular complexes with ß1 and ß4 integrins in primary human keratinocytes and, using immunofluorescence, that these complexes are localized at the plasma membrane, in keeping with a role in adhesion. We confirmed that this protein is a key player of keratinocyte adhesion because in absence of interaction between CD98hc and integrins, ß1 integrin failed to translocate from the cytoplasm to the plasma membrane and keratinocytes expressed epidermal differentiation markers. CONCLUSIONS: All these data strongly suggested that CD98hc is involved in integrin trafficking and by consequence, in keratinocyte adhesion and differentiation.


Subject(s)
Fusion Regulatory Protein 1, Heavy Chain/physiology , Keratinocytes/cytology , Keratinocytes/physiology , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Membrane/physiology , Cells, Cultured , Humans , Integrin beta1/physiology , Integrin beta4/physiology
9.
Biochem Biophys Res Commun ; 397(2): 290-5, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20510671

ABSTRACT

It is known that ozone stress can induce airway hyperresponsiveness (AHR). The underlying cellular and molecular mechanisms are not fully understood. We constructed a successive ozone-stressed rat model and showed that AHR caused by ozone stress presented as increased lung resistance (R(L)) to inhaled histamine but not baseline R(L). Meanwhile, structural disruption and decreased expression of integrin beta4 on airway epithelia were observed. Further regression analysis revealed a significant negative correlation between increases in R(L) to histamine (at 0.32 mg/ml) and mRNA expression of integrin beta4. Moreover, when integrin beta4 on human bronchial epithelial cells was knocked down, we found that reactive oxygen species was increased and apoptosis rates were higher. Overall, this study suggests that downregulation of integrin beta4 is important for the development ozone stress-induced AHR, presumably because it causes increased oxidative damage and epithelial apoptosis.


Subject(s)
Air Pollutants/toxicity , Airway Resistance , Bronchial Hyperreactivity/chemically induced , Integrin beta4/physiology , Oxidative Stress , Ozone/toxicity , Administration, Inhalation , Animals , Apoptosis , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Cell Line , Gene Knockdown Techniques , Histamine/administration & dosage , Humans , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Respiratory Mucosa/physiopathology
10.
J Periodontal Res ; 45(2): 284-91, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20470260

ABSTRACT

BACKGROUND AND OBJECTIVE: It remains controversial whether or not the junctional epithelium cells that are directly attached to teeth migrate on the enamel surface, as those cells are able to adhere firmly to the enamel. The aim of this study was to investigate the expression patterns of laminin gamma(2), integrin beta(4) and integrin alpha(3), and to examine their potential function in cell migration. MATERIAL AND METHODS: Oral epithelium cells obtained from Sprague-Dawley rats were established in primary culture. We employed a wound-healing assay to characterize the direction of cell extension at the start of cell migration, and observed different localizations of laminin and integrins using immunofluorescence. For functional analyses of integrins, we employed a phosphatidylinositol-3-kinase (PI3K) activator to promote integrin beta(4) function and used P1B5 to inhibit integrin alpha(3) function, and we analyzed the percentage of re-epithelialization as the migration function. RESULTS: Marked accumulation of laminin gamma(2) was detected in the peripheral cytoplasm of cells adjacent to the wound area, as shown by the results of the migration assay. Integrin beta(4) was detected in the distal cell processes of actively migrating cells, while integrin alpha(3) was found in cell membranes of cells adjacent to the wound area. In the functional analyses, the percentage of re-epithelialization was significantly lower in the PI3K-activator group and in the P1B5-treated group (2.5% and 7.2%, respectively) than in the control group (39.0%) (p < 0.01). CONCLUSION: The results suggest that laminin gamma(2) is secreted as a foothold for cell migration, that integrin beta(4) participates in cell adhesion and that integrin alpha(3) is involved in cell migration in the primary culture cells.


Subject(s)
Cell Adhesion Molecules/physiology , Epithelial Attachment/cytology , Integrin alpha3/physiology , Integrin beta4/physiology , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/analysis , Cell Membrane/ultrastructure , Cell Movement/physiology , Cell Nucleus/ultrastructure , Cell Surface Extensions/ultrastructure , Cells, Cultured , Coloring Agents , Cytoplasm/ultrastructure , Enzyme Activation , Epithelial Attachment/physiology , Epithelial Cells/physiology , Epithelial Cells/ultrastructure , Fluorescent Antibody Technique , Integrin alpha3/analysis , Integrin alpha3/drug effects , Integrin beta4/analysis , Integrin beta4/drug effects , Microscopy, Confocal , Phosphatidylinositol 3-Kinases/pharmacology , Rats , Rats, Sprague-Dawley , Wound Healing/physiology , Kalinin
11.
Mol Cell Biochem ; 341(1-2): 259-69, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20364299

ABSTRACT

Integrin beta 4 (ITGB4) is a structural adhesion molecule which engages in maintaining the integrity of airway epithelial cells. Its specific cytomembrane structural feature strongly indicates that ITGB4 may engage in many signaling pathways and physiologic processes. However, in addition to adhesion, the specific biologic significance of ITGB4 in airway epithelial cells is almost unknown. In this article, we investigated the expression and functional properties of ITGB4 in airway epithelial cells in vivo and in vitro. Human bronchial epithelial cell line (16HBE14O-cells) and primary rat tracheal epithelial cells (RTE cells) were used to determine ITGB4 expression under ozone tress or mechanical damage, respectively. An ovalbumin (OVA)-challenged asthma model was used to investigate ITGB4 expression after antigen exposure in vivo. In addition, an ITGB4 overexpression vector and ITGB4 silence virus vector were constructed and transfected into RTE cells. Then, wound repair ability and anti-oxidation capacity was evaluated. Our results demonstrated that, on the edge of mechanically wounded cell areas, ITGB4 expression was increased after mechanical injury. After ozone stress, upregulation expression of ITGB4 was also detected. In the OVA-challenged asthma model, ITGB4 expression was decreased on airway epithelial cells accompanying with structural disruption and damage of anti-oxidation capacity. Besides, our study revealed that upregulation of ITGB4 promotes wound repair ability and anti-oxidative ability, while such abilities were blocked when ITGB4 was silenced. Taken together, these results showed that ITGB4 was a new interesting molecule involved in the regulation of wound repair and anti-oxidation processes for airway epithelial cells.


Subject(s)
Antioxidants , Epithelial Cells/physiology , Integrin beta4/physiology , Respiratory System/cytology , Wound Healing , Animals , Asthma/pathology , Bronchi/cytology , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Integrin beta4/biosynthesis , Ozone/pharmacology , Rats , Trachea/cytology
12.
Oncogene ; 28(38): 3401-11, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19597468

ABSTRACT

The development of pulmonary metastasis is the major cause of death in osteosarcoma, and its molecular basis is poorly understood. In this study, we show that beta4 integrin is highly expressed in human osteosarcoma cell lines and tumor samples. Furthermore, highly metastatic MNNG-HOS cells have increased levels of beta4 integrin. Suppression of beta4 integrin expression by shRNA and disruption of beta4 integrin function by transfection of dominant-negative beta4 integrin was sufficient to revert this highly metastatic phenotype in the MNNG-HOS model without significantly affecting primary tumor growth. These findings suggest a role for beta4 integrin expression in the metastatic phenotype in human osteosarcoma cells. In addition, we identified a previously uncharacterized interaction between beta4 integrin and ezrin, a membrane-cytoskeletal linker protein that is implicated in the metastatic behavior of osteosarcoma. The beta4 integrin-ezrin interaction appears to be critical for maintenance of beta4 integrin expression. These data begin to integrate ezrin and beta4 integrin expression into a model of action for the mechanism of osteosarcoma metastases.


Subject(s)
Bone Neoplasms/pathology , Cytoskeletal Proteins/physiology , Integrin beta4/physiology , Osteosarcoma/secondary , Cell Line, Tumor , Humans , Integrin beta4/analysis
13.
J Invest Dermatol ; 129(8): 2031-45, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19212345

ABSTRACT

Flightless I (Flii), a highly conserved member of the gelsolin family of actin-remodelling proteins associates with actin structures and is involved in cellular motility and adhesion. Our previous studies have shown that Flii is an important negative regulator of wound repair. Here, we show that Flii affects hemidesmosome formation and integrin-mediated keratinocyte adhesion and migration. Impaired hemidesmosome formation and sparse arrangements of keratin cytoskeleton tonofilaments and actin cytoskeleton anchoring fibrils were observed in Flii(Tg/+) and Flii(Tg/Tg) mice with their skin being significantly more fragile than Flii(+/-) and WT mice. Flii(+/-) primary keratinocytes showed increased adhesion on laminin and collagen I than WT and Flii(Tg/Tg) primary keratinocytes. Decreased expression of CD151 and laminin-binding integrins alpha3, beta1, alpha6 and beta4 were observed in Flii overexpressing wounds, which could contribute to the impaired wound re-epithelialization observed in these mice. Flii interacts with proteins directly linked to the cytoplasmic domain of integrin receptors suggesting that it may be a mechanical link between ligand-bound integrin receptors and the actin cytoskeleton driving adhesion-signaling pathways. Therefore Flii may regulate wound repair through its effect on hemidesmosome formation and integrin-mediated cellular adhesion and migration.


Subject(s)
Cytoskeletal Proteins/physiology , Hemidesmosomes/physiology , Integrins/physiology , Wound Healing/physiology , Animals , Antigens, CD/analysis , Carrier Proteins , Cell Adhesion , Cell Movement , Cells, Cultured , Cytoskeletal Proteins/genetics , Female , Fibroblasts/physiology , Integrin alpha6/physiology , Integrin beta1/physiology , Integrin beta4/physiology , Keratinocytes/physiology , Laminin/genetics , Mice , Mice, Inbred BALB C , Microfilament Proteins , Signal Transduction , Tetraspanin 24 , Trans-Activators
14.
Int J Biochem Cell Biol ; 41(4): 916-24, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18834954

ABSTRACT

Neural stem cells are capable of differentiating into three major neural cell types, but the underlying molecular mechanisms remain unclear. Here, we investigated the mechanism by which integrin beta4 modulates mouse neural stem cell differentiation in vitro. Inhibition of endogenous integrin beta4 by RNA interference inhibited the cell differentiation and the expression of fibroblast growth factor receptor 2 but not fibroblast growth factor receptor 1 or fibroblast growth factor receptor 3. Overexpression of integrin beta4 in neural stem cells promoted neural stem cell differentiation. Furthermore, integrin beta4-induced differentiation of neural stem cells was attenuated by SU5402, the inhibitor of fibroblast growth factor receptors. Finally, we investigated the role of integrin beta4 in neural stem cell survival: knockdown of integrin beta4 did not affect survival or apoptosis of neural stem cells. These data provide evidence that integrin beta4 promotes differentiation of mouse neural stem cells in vitro possibly through fibroblast growth factor receptor 2.


Subject(s)
Cell Differentiation/physiology , Integrin beta4/physiology , Neurons/cytology , Stem Cells/cytology , Animals , Cell Culture Techniques , Female , Fluorescent Antibody Technique , Gene Expression , Integrin beta4/genetics , Integrin beta4/metabolism , Mice , RNA Interference , RNA, Small Interfering/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Transfection
15.
Cell Commun Adhes ; 15(4): 317-31, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18979297

ABSTRACT

Integrin alpha6beta4-mediated adhesion interactions play key roles in keratinocyte and epithelial tumor cell biology. In order to evaluate how alpha6beta4 adhesion interactions contribute to these important cellular processes, the authors generated soluble versions of the integrin by recombinant expression of the subunit ectodomains fused to a human immunoglobulin G (IgG) Fc constant domain. Coexpression of the appropriate subunits enabled dimerization, secretion and purification of stable Fc-containing alpha6beta4 heterodimers. The soluble proteins exhibited the same metal ion and ligand dependency in their binding characteristics as intact alpha6beta4. Using these reagents in combination with anti-beta4 antibodies, the authors identified two distinct functional epitopes on the beta4 subunit. They demonstrated the involvement of one epitope in adhesion interactions and the other in regulating adhesion-independent growth in alpha6beta4-expressing tumor cell lines. The availability of these soluble integrin reagents and the data provided herein help to further delineate the structure-function relationships regulating alpha6beta4 signaling biology.


Subject(s)
Integrin alpha6beta4/physiology , Integrin beta4/chemistry , Animals , Antibodies/metabolism , CHO Cells , Cell Adhesion , Cell Communication , Cell Line, Tumor , Cricetinae , Cricetulus , Dimerization , Humans , Integrin beta4/immunology , Integrin beta4/physiology , K562 Cells , Keratinocytes/cytology , Keratinocytes/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Signal Transduction , Structure-Activity Relationship
16.
FEBS J ; 275(22): 5725-32, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18959757

ABSTRACT

Integrin beta4 is a tissue-specific protein, but its role in autophagy of lung adenocarcinoma cells is not clear. In this study, we used microtubule-associated protein 1 light chain 3 processing and acridine orange staining to reveal that knockdown of integrin beta4 by its specific siRNA induced autophagic cell death in A549 lung cancer cells. Next, we investigated the effects of siRNA-mediated downregulation of integrin beta4 on cell death and the level of p53. The proportion of dead cells and level of p53 were significantly increased. Inhibition of autophagy by the inhibitor 3-methyladenine attenuated the cell death induced by integrin beta4 knockdown. To further understand the relationship between p53 and integrin beta4 in autophagic cell death, we inhibited the expression of integrin beta4 by its specific siRNA in p53-mutated H322 lung cancer cells. Knockdown of integrin beta4 could not induce autophagic cell death in H322 cells. The data suggest that integrin beta4 is implicated in and associated with p53 in autophagy of lung cancer cells.


Subject(s)
Autophagy , Integrin beta4/physiology , Cell Death , Cell Line, Tumor , Gene Silencing , Humans , Lung Neoplasms , RNA, Small Interfering/pharmacology , Tumor Suppressor Protein p53
17.
J Neurosci ; 28(44): 11292-303, 2008 Oct 29.
Article in English | MEDLINE | ID: mdl-18971471

ABSTRACT

Several different integrins participate in the complex interactions that promote repair of the peripheral nervous system. The role of the integrin alpha6beta4 in peripheral nerve regeneration was investigated in mice by cre-mediated deletion of the Itgb4 (beta4) gene in Schwann cells. After a crush lesion of the sciatic nerve, the recovery of motor, but not that of sensory, nerve function in beta4(-/-) mice was delayed. Immunostaining of neurofilament-200 showed that there also is a significant reduction in the number of newly outgrowing nerve sprouts in beta4(-/-) mice. Morphometric quantitative measurements revealed that fewer axons are myelinated in the nonlesioned beta4(-/-) nerves. After a sciatic nerve crush lesion, beta4(-/-) mice did not only have fewer myelinated axons compared with lesioned wild-type nerve, but their axons also showed a higher g-ratio and a thinner myelin sheath, pointing at reduced myelination. This study revealed that the beta4 protein remains expressed in the early stages of peripheral regeneration, albeit at levels lower than those before the lesion was inflicted, and showed that laminin deposition is not altered in the absence of beta4. These results together demonstrate that integrin alpha6beta4 plays an essential role in axonal regeneration and subsequent myelination.


Subject(s)
Gene Deletion , Integrin beta4/genetics , Nerve Regeneration/physiology , Schwann Cells/physiology , Sciatic Neuropathy/genetics , Animals , Female , Integrin beta4/biosynthesis , Integrin beta4/physiology , Male , Mice , Mice, Transgenic , Nerve Regeneration/genetics , Peripheral Nerves/pathology , Peripheral Nerves/physiology , Schwann Cells/pathology , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Time Factors
18.
Toxicol Appl Pharmacol ; 226(2): 178-91, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17961621

ABSTRACT

Hepatocyte growth factor (HGF) and its receptor, Met, known to control invasive growth program have recently been shown to play crucial roles in the survival of breast cancer patients. The diet-derived flavonoids have been reported to possess anti-invasion properties; however, knowledge on the pharmacological and molecular mechanisms in suppressing HGF/Met-mediated tumor invasion and metastasis is poorly understood. In our preliminary study, we use HGF as an invasive inducer to investigate the effect of flavonoids including apigenin, naringenin, genistein and kaempferol on HGF-dependent invasive growth of MDA-MB-231 human breast cancer cells. Results show that apigenin presents the most potent anti-migration and anti-invasion properties by Boyden chamber assay. Furthermore, apigenin represses the HGF-induced cell motility and scattering and inhibits the HGF-promoted cell migration and invasion in a dose-dependent manner. The effect of apigenin on HGF-induced signaling activation involving invasive growth was evaluated by immunoblotting analysis, it shows that apigenin blocks the HGF-induced Akt phosphorylation but not Met, ERK, and JNK phosphorylation. In addition to MDA-MB-231 cells, apigenin exhibits inhibitory effect on HGF-induced Akt phosphorylation in hepatoma SK-Hep1 cells and lung carcinoma A549 cells. By indirect immunofluorescence microscopy assay, apigenin inhibits the HGF-induced clustering of beta 4 integrin at actin-rich adhesive site and lamellipodia through PI3K-dependent manner. Treatment of apigenin inhibited HGF-stimulated integrin beta 4 function including cell-matrix adhesion and cell-endothelial cells adhesion in MDA-MB-231 cells. By Akt-siRNA transfection analysis, it confirmed that apigenin inhibited HGF-promoted invasive growth involving blocking PI3K/Akt pathway. Finally, we evaluated the effect of apigenin on HGF-promoted metastasis by lung colonization of tumor cells in nude mice and organ metastasis of tumor cells in chick embryo. By histological and gross examination of mouse lung and real-time PCR analysis of human alu in host tissues, it showed that apigenin, wortmannin, as well as anti-beta 4 antibody all inhibit HGF-promoted metastasis. These data support the inhibitory effect of apigenin on HGF-promoted invasive growth and metastasis involving blocking PI3K/Akt pathway and integrin beta 4 function.


Subject(s)
Apigenin/pharmacology , Breast Neoplasms/pathology , Hepatocyte Growth Factor/physiology , Integrin beta4/physiology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chick Embryo , Female , Flavanones/pharmacology , Genistein/pharmacology , Humans , Kaempferols/pharmacology , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis
19.
Int J Biochem Cell Biol ; 40(4): 689-99, 2008.
Article in English | MEDLINE | ID: mdl-18006359

ABSTRACT

Recently, the specific roles of integrin beta4 in the signaling networks that drive pathological angiogenesis and tumor progression have been revealed. Our previous study showed that integrin beta4 might be involved in neuron survival signal transduction. To further our study on the role of integrin beta4 in the survival and apoptosis of primary cultured mouse neurons, we inhibited the expression of integrin beta4 by its specific small interfering RNA. Viability of the cells remarkably declined, and neurons underwent apoptosis with down-regulation of integrin beta4. Next, we investigated the effect of siRNA-mediated down-regulation of integrin beta4 on the level of intracellular reactive oxygen species and the activities of NADPH oxidase and superoxide dismutase. The level of reactive oxygen species in the neurons was elevated significantly, the activities of manganese-dependent superoxide dismutase and copper/zinc-dependent superoxide dismutase were not altered, but the activity of NADPH oxidase was increased. Furthermore, inhibition of NADPH oxidase by its specific inhibitor dibenziodolium chloride attenuated the neuronal death induced by integrin beta4 knockdown. The data suggest that integrin beta4 is a key factor in neuron survival and apoptosis and indicate that this integrin subunit might perform its action through regulating NADPH oxidase and the level of reactive oxygen species in neuronal survival and apoptosis.


Subject(s)
Apoptosis/physiology , Integrin beta4/physiology , NADPH Oxidases/metabolism , Neurons/metabolism , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Integrin beta4/genetics , Integrin beta4/metabolism , Mice , Neurons/cytology , RNA Interference
20.
Trends Pharmacol Sci ; 28(10): 506-11, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17822782

ABSTRACT

The integrins play key roles in the signaling networks that drive pathological angiogenesis and tumor progression. Integrin beta4 is a laminin receptor upregulated in tumor cells and angiogenic endothelial cells. Biochemical studies have indicated that beta4 combines with and enhances the signaling function of multiple receptor tyrosine kinases, including ErbB2, EGF-R and Met. Genetic studies have revealed that beta4 signaling promotes both angiogenesis and tumorigenesis. Here, I discuss the hypothesis that beta4 promotes both processes by amplifying receptor-tyrosine-kinase signaling. Therefore, I propose that a simultaneous blockade of beta4 and receptor-tyrosine-kinase signaling represents a rational approach to cancer and anti-angiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Integrin beta4/drug effects , Animals , ErbB Receptors/physiology , Humans , Integrin beta4/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-met , Receptor, ErbB-2/physiology , Receptors, Growth Factor/physiology , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...