Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters











Publication year range
1.
Am J Physiol Cell Physiol ; 326(5): C1308-C1319, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38497112

ABSTRACT

Integrin receptors for the extracellular matrix activate intracellular signaling pathways that are critical for tissue development, homeostasis, and regeneration/repair, and their loss or dysregulation contributes to many developmental defects and tissue pathologies. This review will focus on tissue remodeling roles for integrin α3ß1, a receptor for laminins found in the basement membranes (BMs) that underlie epithelial cell layers. As a paradigm, we will discuss literature that supports a role for α3ß1 in promoting ability of epidermal keratinocytes to modify their tissue microenvironment during skin development, wound healing, or tumorigenesis. Preclinical and clinical studies have shown that this role depends largely on ability of α3ß1 to govern the keratinocyte's repertoire of secreted proteins, or the "secretome," including 1) matrix proteins and proteases involved in matrix remodeling and 2) paracrine-acting growth factors/cytokines that stimulate other cells with important tissue remodeling functions (e.g., endothelial cells, fibroblasts, inflammatory cells). Moreover, α3ß1 signaling controls gene expression that helps epithelial cells carry out these functions, including genes that encode secreted matrix proteins, proteases, growth factors, or cytokines. We will review what is known about α3ß1-dependent gene regulation through both transcription and posttranscriptional mRNA stability. Regarding the latter, we will discuss examples of α3ß1-dependent alternative splicing (AS) or alternative polyadenylation (APA) that prevents inclusion of cis-acting mRNA sequences that would otherwise target the transcript for degradation via nonsense-mediated decay or destabilizing AU-rich elements (AREs) in the 3'-untranslated region (3'-UTR). Finally, we will discuss prospects and anticipated challenges of exploiting α3ß1 as a clinical target for the treatment of cancer or wound healing.


Subject(s)
Endothelial Cells , Integrin alpha3beta1 , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Endothelial Cells/metabolism , Keratinocytes/metabolism , Peptide Hydrolases/metabolism , Cytokines/metabolism , Cell Adhesion
2.
Matrix Biol ; 127: 48-56, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38340968

ABSTRACT

The development of wound therapy targeting integrins is hampered by inadequate understanding of integrin function in cutaneous wound healing and the wound microenvironment. Following cutaneous injury, keratinocytes migrate to restore the skin barrier, and macrophages aid in debris clearance. Thus, both keratinocytes and macrophages are critical to the coordination of tissue repair. Keratinocyte integrins have been shown to participate in this coordinated effort by regulating secreted factors, some of which crosstalk to distinct cells in the wound microenvironment. Epidermal integrin α3ß1 is a receptor for laminin-332 in the cutaneous basement membrane. Here we show that wounds deficient in epidermal α3ß1 express less epidermal-derived macrophage colony-stimulating factor 1 (CSF-1), the primary macrophage-stimulating growth factor. α3ß1-deficient wounds also have fewer wound-proximal macrophages, suggesting that keratinocyte α3ß1 may stimulate wound macrophages through the regulation of CSF-1. Indeed, using a set of immortalized keratinocytes, we demonstrate that keratinocyte-derived CSF-1 supports macrophage growth, and that α3ß1 regulates Csf1 expression through Src-dependent stimulation of Yes-associated protein (YAP)-Transcriptional enhanced associate domain (TEAD)-mediated transcription. Consistently, α3ß1-deficient wounds in vivo display a substantially reduced number of keratinocytes with YAP-positive nuclei. Overall, our current findings identify a novel role for epidermal integrin α3ß1 in regulating the cutaneous wound microenvironment by mediating paracrine crosstalk from keratinocytes to wound macrophages, implicating α3ß1 as a potential target of wound therapy.


Subject(s)
Integrin alpha3beta1 , Macrophage Colony-Stimulating Factor , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Keratinocytes/metabolism , Epidermis , Wound Healing/physiology
3.
Toxins (Basel) ; 15(12)2023 12 14.
Article in English | MEDLINE | ID: mdl-38133204

ABSTRACT

Podocyte dysfunction plays a crucial role in renal injury and is identified as a key contributor to proteinuria in Fabry disease (FD), primarily impacting glomerular filtration function (GFF). The α3ß1 integrins are important for podocyte adhesion to the glomerular basement membrane, and disturbances in these integrins can lead to podocyte injury. Therefore, this study aimed to assess the effects of chloroquine (CQ) on podocytes, as this drug can be used to obtain an in vitro condition analogous to the FD. Murine podocytes were employed in our experiments. The results revealed a dose-dependent reduction in cell viability. CQ at a sub-lethal concentration (1.0 µg/mL) induced lysosomal accumulation significantly (p < 0.0001). Morphological changes were evident through scanning electron microscopy and immunofluorescence, highlighting alterations in F-actin and nucleus morphology. No significant changes were observed in the gene expression of α3ß1 integrins via RT-qPCR. Protein expression of α3 integrin was evaluated with Western Blotting and immunofluorescence, demonstrating its lower detection in podocytes exposed to CQ. Our findings propose a novel in vitro model for exploring secondary Fabry nephropathy, indicating a modulation of α3ß1 integrin and morphological alterations in podocytes under the influence of CQ.


Subject(s)
Fabry Disease , Integrin alpha3beta1 , Kidney Diseases , Podocytes , Animals , Mice , Fabry Disease/metabolism , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Kidney Diseases/metabolism , Podocytes/metabolism , Renal Insufficiency
4.
Cells ; 11(5)2022 02 26.
Article in English | MEDLINE | ID: mdl-35269439

ABSTRACT

Endothelial cells engage extracellular matrix and basement membrane components through integrin-mediated adhesion to promote angiogenesis. Angiogenesis involves the sprouting of endothelial cells from pre-existing vessels, their migration into surrounding tissue, the upregulation of angiogenesis-associated genes, and the formation of new endothelial tubes. To determine whether the endothelial laminin-binding integrins, α6ß4, and α3ß1 contribute to these processes, we employed RNAi technology in organotypic angiogenesis assays, as well in migration assays, in vitro. The endothelial depletion of either α6ß4 or α3ß1 inhibited endothelial sprouting, indicating that these integrins have non-redundant roles in this process. Interestingly, these phenotypes were accompanied by overlapping and distinct changes in the expression of angiogenesis-associated genes. Lastly, depletion of α6ß4, but not α3ß1, inhibited migration. Taken together, these results suggest that laminin-binding integrins regulate processes associated with angiogenesis by distinct and overlapping mechanisms.


Subject(s)
Integrin alpha6beta4 , Laminin , Cell Adhesion/physiology , Endothelial Cells/metabolism , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Integrin alpha6beta4/genetics , Integrin alpha6beta4/metabolism , Laminin/metabolism
5.
PLoS One ; 16(7): e0254714, 2021.
Article in English | MEDLINE | ID: mdl-34270616

ABSTRACT

Integrin receptors for the extracellular matrix play critical roles at all stages of carcinogenesis, including tumor growth, tumor progression and metastasis. The laminin-binding integrin α3ß1 is expressed in all epithelial tissues where it has important roles in cell survival, migration, proliferation, and gene expression programs during normal and pathological tissue remodeling. α3ß1 signaling and adhesion functions promote tumor growth and metastasis in a number of different types of cancer cells. Previously, we used RNA interference (RNAi) technology to suppress the expression of the ITGA3 gene (encoding the α3 subunit) in the triple-negative breast cancer cell line, MDA-MB-231, thereby generating variants of this line with reduced expression of integrin α3ß1. This approach revealed that α3ß1 promotes pro-tumorigenic functions such as cell invasion, lung metastasis, and gene regulation. In the current study, we used CRISPR technology to knock out the ITGA3 gene in MDA-MB-231 cells, thereby ablating expression of integrin α3ß1 entirely. RNA-seq analysis revealed that while the global transcriptome was altered substantially by RNAi-mediated suppression of α3ß1, it was largely unaffected following CRISPR-mediated ablation of α3ß1. Moreover, restoring α3ß1 to the latter cells through inducible expression of α3 cDNA failed to alter gene expression substantially, suggesting that use of CRISPR to abolish α3ß1 led to a decoupling of the integrin from its ability to regulate the transcriptome. Interestingly, both cell invasion in vitro and metastatic colonization in vivo were reduced when α3ß1 was abolished using CRISPR, as we observed previously using RNAi to suppress α3ß1. Taken together, our results show that pro-invasive/pro-metastatic roles for α3ß1 are not dependent on its ability to regulate the transcriptome. Moreover, our finding that use of RNAi versus CRISPR to target α3ß1 produced distinct effects on gene expression underlines the importance of using multiple approaches to obtain a complete picture of an integrin's functions in cancer cells.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Integrin alpha3beta1/genetics , Lung Neoplasms/genetics , Triple Negative Breast Neoplasms/genetics , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Datasets as Topic , Female , Gene Editing , Humans , Lung Neoplasms/secondary , Mice , Neoplasm Invasiveness/genetics , RNA Interference , RNA-Seq , Transcriptome/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
J Invest Dermatol ; 141(4): 732-741.e6, 2021 04.
Article in English | MEDLINE | ID: mdl-32805217

ABSTRACT

Integrin α3ß1 plays a crucial role in tumor formation in the two-stage chemical carcinogenesis model (DMBA and TPA treatment). However, the mechanisms whereby the expression of α3ß1 influences key oncogenic drivers of this established model are not known yet. Using an in vivo mouse model with epidermal deletion of α3ß1 and in vitro Matrigel cultures of transformed keratinocytes, we demonstrate the central role of α3ß1 in promoting the activation of several protumorigenic signaling pathways during the initiation of DMBA/TPA‒driven tumorigenesis. In transformed keratinocytes, α3ß1-mediated focal adhesion kinase/Src activation leads to in vitro growth of spheroids and to strong Akt and STAT 3 activation when the α3ß1-binding partner tetraspanin CD151 is present to stabilize cell‒cell adhesion and promote Smad2 phosphorylation. Remarkably, α3ß1 and CD151 can support Akt and STAT 3 activity independently of α3ß1 ligation by laminin-332 and as such control the essential survival signals required for suprabasal keratin-10 expression during keratinocyte differentiation. These data demonstrate that α3ß1 together with CD151 regulate the signaling pathways that control the survival of differentiating keratinocytes and provide a mechanistic understanding of the essential role of α3ß1 in early stages of skin cancer development.


Subject(s)
Cell Transformation, Neoplastic/pathology , Integrin alpha3beta1/metabolism , Keratinocytes/pathology , Neoplasms, Experimental/pathology , Skin Neoplasms/pathology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Carcinogens/toxicity , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Line , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Epidermis/drug effects , Epidermis/pathology , Humans , Integrin alpha3beta1/genetics , Keratinocytes/drug effects , Mice , Neoplasms, Experimental/chemically induced , Signal Transduction , Skin Neoplasms/chemically induced , Spheroids, Cellular , Tetradecanoylphorbol Acetate/toxicity , Tetraspanin 24/metabolism , Kalinin
7.
Aging (Albany NY) ; 12(23): 24345-24356, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33260159

ABSTRACT

Downregulation of integrins α3ß1 and α5ß1 strongly decreased cell colony formation and in vitro invasion and markedly enhanced anoikis in SK-Mel-147 human melanoma cells. These modifications were accompanied by a marked increase in the levels of active Akt protein kinase, which indicated it played a non-canonical function in the melanoma cells. Pharmacological inhibition of Akt1, an Akt isozyme, in cells depleted of α3ß1 or α5ß1 restored their invasive activity, while inhibition of the Akt 2 isoform did not cause a visible effect. Similar to our previous results with the α2ß1 integrin, this finding suggested that in signaling pathways initiated by α3ß1 and α5ß1, the Akt1 isoform performs a non-canonical function in regulating invasive phenotype of melanoma cells. In contrast, when the effects of Akt inhibitors on anoikis of the melanoma cells were compared, the Akt2 isoform demonstrated a non-canonical activity in which Akt2 suppression led to a significant attenuation of apoptosis in cells with downregulated α3ß1 or α5ß1. Our results were the first evidence that, in the same tumor cells, different integrins can control various manifestations of tumor progression through distinct signaling pathways that are both common to various integrins and specific to a particular receptor.


Subject(s)
Anoikis , Cell Movement , Integrin alpha3beta1/metabolism , Integrin alpha5beta1/metabolism , Melanoma/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/enzymology , Anoikis/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha3beta1/genetics , Integrin alpha5beta1/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology
8.
Life Sci Alliance ; 3(7)2020 07.
Article in English | MEDLINE | ID: mdl-32423907

ABSTRACT

Epidermal-specific deletion of integrin α3ß1 almost completely prevents the formation of papillomas during 7,12-Dimethylbenz[ a ]anthracene/12- O -tetradecanoylphorbol-13-acetate (DMBA/TPA) two-stage skin carcinogenesis. This dramatic decrease in tumorigenesis was thought to be due to an egress and premature differentiation of α3ß1-depleted hair bulge (HB) stem cells (SCs), previously considered to be the cancer cells-of-origin in the DMBA/TPA model. Using a reporter mouse line with inducible deletion of α3ß1 in HBs, we show that HB SCs remain confined to their niche regardless of the presence of α3ß1 and are largely absent from skin tumors. However, tumor formation was significantly decreased in mice deficient for α3ß1 in HB SCs. RNA sequencing of HB SCs isolated from short-term DMBA/TPA-treated skin showed α3ß1-dependent expression of the matricellular protein connective tissue growth factor (CCN2), which was confirmed in vitro, where CCN2 promoted colony formation and 3D growth of transformed keratinocytes. Together, these findings show that HBs contribute to skin tumorigenesis in an α3ß1-dependent manner and suggest a role of HB SCs in creating a permissive environment for tumor growth through the modulation of CCN2 secretion.


Subject(s)
Connective Tissue Growth Factor/genetics , Gene Expression Regulation , Hair Follicle/cytology , Integrin alpha3beta1/metabolism , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Stem Cells/metabolism , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Epidermis/metabolism , Epidermis/pathology , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Immunophenotyping , Integrin alpha3beta1/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Knockout , Neoplasm Staging , Skin Neoplasms/pathology
9.
ACS Appl Mater Interfaces ; 12(13): 14905-14913, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32148016

ABSTRACT

Docetaxel (DTX) widely used for treating nonsmall cell lung cancer (NSCLC) patients is associated with dose-limiting side effects, especially neurotoxicity and myelosuppression. Here, we have developed cyclic cNGQGEQc peptide-directed polymersomal docetaxel (cNGQ-PS-DTX) as a targeted and multifunctional formulation for NSCLC. cNGQ-PS-DTX carrying 8.1 wt % DTX had a size of 93 nm, neutral surface charge, high stability, and glutathione-triggered DTX release behavior. Cytotoxicity studies demonstrated a clearly better antitumor activity of cNGQ-PS-DTX in α3ß1 integrin overexpressing A549 human lung cancer cells than free DTX and nontargeted PS-DTX. cNGQ-PS-DTX showed a remarkably high tolerability (over 8 times better than free DTX) and slow elimination in mice. Importantly, cNGQ-PS-DTX exhibited greatly improved tumor accumulation and higher suppression of subcutaneous and orthotopic A549 xenografts as compared to PS-DTX and free DTX controls. α3ß1 integrin-targeting polymersomal docetaxel emerges as an advanced nanotherapeutic for NSCLC treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Docetaxel/chemistry , Integrin alpha3beta1/metabolism , Lung Neoplasms/drug therapy , A549 Cells , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Docetaxel/pharmacokinetics , Docetaxel/therapeutic use , Drug Carriers/chemistry , Half-Life , Humans , Integrin alpha3beta1/antagonists & inhibitors , Integrin alpha3beta1/genetics , Mice , Mice, Nude , Nanoparticles/chemistry , Peptides/chemistry , Peptides/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
10.
J Hematol Oncol ; 12(1): 56, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182116

ABSTRACT

BACKGROUND: α3ß1 integrin is a promising cancer biomarker and drug target. We previously identified a 9-amino-acid cyclic peptide LXY30 for detecting α3ß1 integrin on the surface of live tumor cells. This study was undertaken to characterize LXY30 in the detection, cellular function, imaging, and targeted delivery of in vitro and in vivo non-small cell lung cancer (NSCLC) models. METHODS: The whole-cell binding assay was performed by incubating NSCLC cells, extracellular vesicles (EVs), and peripheral blood mononuclear cells (PBMCs) with TentaGel resin beads coated with LXY30. In this study, we defined the nanosize EVs as exosomes, which were characterized by flow cytometry, transmission electron microscopy, dynamic light scattering, and Western blots. The function of LXY30 was determined by modulating the epidermal growth factor receptor (EGFR) signaling pathway by growth inhibition and Western blots. For in vivo biodistribution, mice bearing subcutaneous and intracranial NSCLC xenograft tumors were administrated intraveneously with LXY30-biotin/streptavidin-Cy5.5 complex and then analyzed for in vivo and ex vivo optical imaging and histopathology. RESULTS: We showed that LXY30 specifically and sensitively detected α3ß1 integrin-expressing NSCLC cells and tumor-derived exosomes. Tumor DNA isolated from LXY30-enriched plasma exosomes might be used to detect driver oncogenic mutations in patients with metastatic NSCLC. LXY30 only enriches tumor cells but not neutrophils, macrophages, or monocytes in the malignant pleural effusion of NSCLC patients for detecting genomic alterations by next-generation sequencing. LXY30 detected increased α3ß1 integrin expression on the EGFR-mutant NSCLC cells with acquired resistance to erlotinib compared to parental erlotinib-sensitive EGFR-mutant NSCLC cells. We further showed that LXY30 modulated the EGFR signaling pathway independently from another peptide ligand LXW64 targeting αvß3 integrin in erlotinib-resistant, EGFR-mutant H1975 cells. Analysis of The Cancer Genome Atlas (TCGA) revealed high α3 integrin expression was associated with poor prognosis in lung squamous cell carcinoma. LXY30-biotin/streptavidin-Cy5.5 complex had higher uptakes in the subcutaneous and intracranial xenografts of various α3ß1 integrin-expressing lung adenocarcinoma and patient-derived lung squamous cell carcinoma xenografts while sparing the surrounding normal tissues. CONCLUSION: LXY30 is a promising peptide for the cancer diagnosis and in vivo targeted delivery of imaging agents and cancer drugs in NSCLC, independent of histology and tumor genotype.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Integrin alpha3beta1/genetics , Lung Neoplasms/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Disease Models, Animal , Female , Humans , Ligands , Lung Neoplasms/pathology , Mice , Mice, Nude , Peptides
11.
J Biol Chem ; 294(12): 4425-4436, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30659093

ABSTRACT

Aberrant N-glycan sialylation of glycoproteins is closely associated with malignant phenotypes of cancer cells and metastatic potential, which includes cell adhesion, migration, and growth. Recently, phosphatidylinositol 4-kinase IIα (PI4KIIα), which is localized to the trans-Golgi network, was identified as a regulator of Golgi phosphoprotein 3 (GOLPH3) and of vesicle transport in the Golgi apparatus. GOLPH3 is a target of PI4KIIα and helps anchor sialyltransferases and thereby regulates sialylation of cell surface receptors. However, how PI4KIIα-mediated sialyation of cell surface proteins is regulated remains unclear. In this study, using several cell lines, CRISPR/Cas9-based gene knockout and short hairpin RNA-mediated silencing, RT-PCR, lentivirus-mediated overexpression, and immunoblotting methods, we confirmed that PI4KIIα knockdown suppresses the sialylation of N-glycans on the cell surface, in Akt phosphorylation and activation, and integrin α3-mediated cell migration of MDA-MB-231 breast cancer cells. Interestingly, both integrin α3ß1 and PI4KIIα co-localized to the trans-Golgi network, where they physically interacted with each other, and PI4KIIα specifically associated with integrin α3 but not α5. Furthermore, overexpression of both integrin α3ß1 and PI4KIIα induced hypersialylation. Conversely, integrin α3 knockout significantly inhibited the sialylation of membrane proteins, such as the epidermal growth factor receptor, as well as in total cell lysates. These findings suggest that the malignant phenotype of cancer cells is affected by a sialylation mechanism that is regulated by a complex between PI4KIIα and integrin α3ß1.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Integrin alpha3beta1/metabolism , N-Acetylneuraminic Acid/metabolism , 1-Phosphatidylinositol 4-Kinase/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Cell Movement , Gene Knockdown Techniques , Humans , Integrin alpha3beta1/genetics , Membrane Proteins/metabolism , Phosphorylation , Polysaccharides/metabolism , Protein Binding , Signal Transduction , trans-Golgi Network/metabolism
12.
Matrix Biol ; 77: 101-116, 2019 04.
Article in English | MEDLINE | ID: mdl-30193894

ABSTRACT

Integrins, the major receptors for cell-extracellular matrix (ECM) interactions, regulate multiple cell biological processes including adhesion, migration, proliferation and growth factor-dependent signaling. The principal laminin (LM) binding integrins α3ß1, α6ß1 and α6ß4 are usually co-expressed in cells and bind to multiple laminins with different affinities making it difficult to define their specific function. In this study, we generated kidney epithelial collecting duct (CD) cells that lack both the α3 and α6 integrin subunits. This deletion impaired cell adhesion and migration to LM-332 and LM-511 more than deleting α3 or α6 alone. Cell adhesion mediated by both α3ß1 and α6 integrins was PI3K independent, but required K63-linked polyubiquitination of Akt by the ubiquitin-modifying enzyme TRAF6. Moreover, we provide evidence that glial-derived neurotrophic factor (GDNF) and fibroblast growth factor 10 (FGF10)- mediated cell signaling, spreading and proliferation were severely compromised in double integrin α3/α6- but not single α3- or α6-null CD cells. Interestingly, these growth factor-dependent cell functions required both PI3K- and TRAF6-dependent Akt activation. These data suggest that expression of the integrin α3 or α6 subunit is sufficient to mediate GDNF- and FGF10-dependent spreading, proliferation and signaling on LM-511. Thus, our study shows that α3 and α6 containing integrins promote distinct functions and signaling by CD cells on laminin substrata.


Subject(s)
Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Integrin alpha3/metabolism , Integrin alpha6/metabolism , Laminin/metabolism , Signal Transduction , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects , Fibroblast Growth Factor 10/pharmacology , Gene Deletion , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Integrin alpha3/genetics , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Integrin alpha6/genetics , Integrin alpha6beta1/genetics , Integrin alpha6beta1/metabolism , Integrin alpha6beta4/genetics , Integrin alpha6beta4/metabolism , Intracellular Signaling Peptides and Proteins , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Laminin/chemistry , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Kalinin
13.
Asian Pac J Cancer Prev ; 19(9): 2493-2498, 2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30256042

ABSTRACT

Objective: To investigate the regulatory role of fibronectin (FN) in the formation of multicellular aggregate (MCA) in ovarian cancer SKOV3 and OVCAR-3 cells and integrin expression. Methods: The dynamic formation of MCA in SKOV3 and OVCAR-3 was determined using the liquid overlay technique in the presence or absence of FN, anti-FN, RGD peptide, control RGE. The expression of α3ß1, α4ß1 and α5ß1 integrin in monolayer cells, MCA and FN-treated MCA were determined by flow cytometry and quantitative RT-PCR. Results: OVCAR-3 and SKOV3 MCA were formed on the 4th and 8th day and peaked on the 6th and 9th day, respectively. Treatment with different concentrations of FN, LN, type IV collagen and control RGE peptide promoted MCA growth, which was mitigated by anti-FN and RGD peptide. In comparison with monolayer cells, up-regulated α3ß1, α4ß1 and α5ß1 expression were detected in MCA while treatment with FN in both cells. Conclusions: OVCAR-3 and SKOV3 cells had varying dynamic formation of MCA in our experimental system. FN enhanced MCA formation in both cells, which was associated with increased expression of 3ß1, α4ß1 and α5ß1 in the MCA. Therefore, FN and these integrins may be new therapeutic targets for intervention of ovarian cancer metastasis.


Subject(s)
Fibronectins/pharmacology , Gene Expression Regulation/drug effects , Integrin alpha3beta1/metabolism , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Ovarian Neoplasms/pathology , Protein Aggregates/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Humans , Integrin alpha3beta1/genetics , Integrin alpha4beta1/genetics , Integrin alpha5beta1/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured
14.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29899108

ABSTRACT

Host receptor usage by Kaposi's sarcoma-associated herpesvirus (KSHV) has been best studied using primary microvascular endothelial and fibroblast cells, although the virus infects a wide variety of cell types in culture and in natural infections. In these two infection models, KSHV adheres to the cell though heparan sulfate (HS) binding and then interacts with a complex of EphA2, xCT, and integrins α3ß1, αVß3, and αVß5 to catalyze viral entry. We dissected this receptor complex at the genetic level with CRISPR-Cas9 to precisely determine receptor usage in two epithelial cell lines. Surprisingly, we discovered an infection mechanism that requires HS and EphA2 but is independent of αV- and ß1-family integrin expression. Furthermore, infection appears to be independent of the EphA2 intracellular domain. We also demonstrated that while two other endogenous Eph receptors were dispensable for KSHV infection, transduced EphA4 and EphA5 significantly enhanced infection of cells lacking EphA2.IMPORTANCE Our data reveal an integrin-independent route of KSHV infection and suggest that multiple Eph receptors besides EphA2 can promote and regulate infection. Since integrins and Eph receptors are large protein families with diverse expression patterns across cells and tissues, we propose that KSHV may engage with several proteins from both families in different combinations to negotiate successful entry into diverse cell types.


Subject(s)
Herpesviridae Infections/virology , Herpesvirus 8, Human , Integrin alpha3beta1/genetics , Integrin alphaVbeta3/genetics , Receptors, Vitronectin/genetics , Virus Internalization , CRISPR-Cas Systems , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Endothelial Cells/virology , Ephrin-A2/genetics , Fibroblasts/virology , Gene Editing , Gene Expression Regulation, Viral , HeLa Cells , Herpesvirus 8, Human/physiology , Humans , Integrin alpha3beta1/metabolism , Integrin alphaVbeta3/metabolism , Pinocytosis , Receptor, EphA2 , Receptors, Vitronectin/metabolism , Signal Transduction/genetics
15.
J Cell Sci ; 131(10)2018 05 22.
Article in English | MEDLINE | ID: mdl-29700202

ABSTRACT

Structural analyses of ß2 and ß3 integrins have revealed that they generally assume a compact bent conformation in the resting state and undergo a global conformational transition involving extension during upregulation of ligand affinity, collectively called the 'switchblade model'. This hypothesis, however, has not been extensively tested for other classes of integrins. We prepared a set of recombinant integrin ectodomain fragments including αvß3, α2ß1, α3ß1, α5ß1, α6ß1 and α6ß4, and used negative-stain electron microscopy to examine their structures under various conditions. In contrast to αvß3 integrin, which exhibited a severely bent conformation in low-affinity 5 mM Ca2+ conditions, all ß1 integrin heterodimers displayed a mixed population of half-bent to fully extended conformations. Moreover, they did not undergo significant conformational change upon activation by Mn2+ Integrin α6ß4 was even more resistant to conformational regulation, showing a completely extended structure regardless of the buffer conditions. These results suggest that the mechanisms of conformational regulation of integrins are more diverse and complex than previously thought, requiring more experimental scrutiny for each integrin subfamily member.


Subject(s)
Integrin alpha6beta4/chemistry , Integrin beta1/chemistry , Integrin beta4/chemistry , Calcium/chemistry , Calcium/metabolism , Cell Line , Humans , Integrin alpha3beta1/chemistry , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Integrin alpha6beta4/genetics , Integrin alpha6beta4/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Integrin beta4/genetics , Integrin beta4/metabolism , Ligands , Microscopy, Electron , Protein Conformation , Protein Domains
16.
J Invest Dermatol ; 138(2): 444-451, 2018 02.
Article in English | MEDLINE | ID: mdl-28923241

ABSTRACT

Proteolytic processing of the laminin-γ2 chain is a hallmark of basement membrane maturation in the skin. Integrin α3ß1, a major receptor for epidermal adhesion to laminin-332, is critical for proper basement membrane organization during skin development and wound healing. Previously, we identified a role for α3ß1 in promoting the processing of laminin-γ2 in cultured keratinocytes in vitro and in wound epidermis in vivo. In this study we identify the Bmp1 gene, which encodes variants of the mTLD/BMP-1 metalloproteases, as a critical regulator of α3ß1-dependent laminin-γ2 processing, thereby expanding the role of this integrin in controlling the secretion by the epidermis of factors that modulate the tissue microenvironment. Because our previous studies identified another epidermal integrin, α9ß1, as a suppressive regulator of α3ß1-dependent wound angiogenesis, we investigated whether α9ß1 has a similar cross-suppressive effect on the ability of α3ß1 to promote basement membrane organization. Here, we show that, rather than a cross-suppressive role, α9ß1 has an opposing role in basement membrane assembly/maturation through reduced laminin-γ2 processing via mTLD/BMP-1. Although α3ß1 promotes this process during wound healing, α9ß1 has an inhibitory role, suggesting that regulation of basement membrane assembly requires a complex interplay between these distinct epidermal integrins.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Integrin alpha3beta1/metabolism , Integrins/metabolism , Laminin/metabolism , Wound Healing/physiology , Wounds and Injuries/pathology , Animals , Basement Membrane/metabolism , Bone Morphogenetic Protein 1/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Disease Models, Animal , Epidermis/injuries , Epidermis/metabolism , Humans , Integrin alpha3beta1/genetics , Integrins/genetics , Keratinocytes , Mice , Mice, Knockout , Proteolysis , RNA, Small Interfering/metabolism , Wounds and Injuries/etiology , Kalinin
17.
Eur J Cell Biol ; 96(3): 266-275, 2017 May.
Article in English | MEDLINE | ID: mdl-28336087

ABSTRACT

Dermatopontin (DPT) is a matricellular protein with cardinal roles in cutaneous wound healing. The protein is also reported to be altered in various anomalies including cancer. The present study is aimed to unravel the role of DPT in angiogenesis which is imperative in many physiological and pathological processes. DPT's capabilities on promoting angiogenesis were assessed using various in vitro and ex vivo systems. The results indicated that DPT enhances cell motility and induces lamellipodia formation in endothelial cells. Additionally, we noticed that DPT stimulates tube formation in endothelial cells when plated on a matrigel substrate. However, it was observed that DPT had no effect on the proliferation of endothelial cells even at higher concentrations and prolonged treatment periods. Additional experiments on CAM and aortic arch assays apparently depicted that DPT promotes neovascularisation and tube sprouting, thus unraveling its prominent role in angiogenesis. Further, PCR analysis revealed that endothelial cells are devoid of DPT expression, but when exogenously supplied, modulates the expression of transforming growth factor ß1 and integrin α3ß1 which are reported to have crucial roles in endothelial cell behaviour during angiogenesis. In conclusion, DPT possess vital pro-angiogenic properties and thus retains promising therapeutic values in managing chronic wounds and cancer.


Subject(s)
Chondroitin Sulfate Proteoglycans/pharmacology , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Extracellular Matrix Proteins/pharmacology , Integrin alpha3beta1/metabolism , Neovascularization, Physiologic , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Chick Embryo , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Integrin alpha3beta1/genetics , Recombinant Proteins , Transforming Growth Factor beta/genetics
18.
Cell Physiol Biochem ; 41(2): 689-700, 2017.
Article in English | MEDLINE | ID: mdl-28214836

ABSTRACT

BACKGROUND/AIMS: Hepatitis B virus (HBV)-associated glomerulonephritis (HBV-GN) is characterized by a reduced number of podocytes due to apoptosis and shedding from the basement membrane. However, the pathological mechanism of HBV-GN is unclear. We previously showed that hepatitis B virus X protein (HBx) promotes apoptosis in tubular epithelial cells. In this study, we transfected podocytes with HBx and examined the effects on adhesion and apoptosis of these cells. METHODS: Podocytes were transfected with pc-DNA3.1 (+)-HBx. One control group was not transfected and another control group was transfected with empty plasmids. Podocyte adhesion was assessed by a fluorescence assay, apoptosis was measured by flow cytometry and fluorescence microscopy, and expression of α3ß1 integrin was determined by western blotting and the reverse transcription polymerase chain reaction (RT-PCR). Activity of caspase-8 was measured by a spectrophotometric assay. RESULTS: Relative to controls, podocytes with pc-DNA3.1(+)-HBx had reduced cell adhesion, increased apoptosis, reduced expression of α3ß1 integrin, and increased caspase-8 activity. ß1 integrin blockage reduced podocyte adhesion, but increased apoptosis and caspase-8 activity. Treatment of transfected podocytes with a caspase-8 inhibitor (Z-IETD-FMK) had no effect on the HBx-mediated integrin downregulation and reduced podocyte adhesion, suggesting that α3ß1 integrin downregulaton is sufficient to alter cell adhesion. CONCLUSIONS: Our in vitro results indicate that HBx reduced podocyte adhesion and expression of α3ß1 integrin, and increased apoptosis. Moreover, HBx-mediated downregulation of α3ß1 integrin expression is sufficient to reduce podocyte adhesion. HBx-induced apoptosis of podocytes may contribute to HBV-GN.


Subject(s)
Trans-Activators/metabolism , A549 Cells , Animals , Antibodies/immunology , Apoptosis , Caspase 8/analysis , Caspase 8/chemistry , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Cell Adhesion/drug effects , Cell Line , Down-Regulation , Humans , Integrin alpha3beta1/genetics , Integrin alpha3beta1/immunology , Integrin alpha3beta1/metabolism , Mice , Oligopeptides/pharmacology , Plasmids/metabolism , Spectrophotometry , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
19.
Matrix Biol ; 57-58: 213-243, 2017 01.
Article in English | MEDLINE | ID: mdl-27562932

ABSTRACT

Integrins play an important role in cell adhesion by linking the cytoskeleton of cells to components in the extracellular matrix. In this capacity, integrins cooperate with different cell surface receptors, including growth factor receptors and G-protein coupled receptors, to regulate intracellular signaling pathways that control cell polarization, spreading, migration, survival, and gene expression. A distinct subfamily of molecules in the integrin family of adhesion receptors is formed by receptors that mediate cell adhesion to laminins, major components of the basement membrane that lie under clusters of cells or surround them, separating them from other cells and/or adjacent connective tissue. During the past decades, many studies have provided evidence for a role of laminin-binding integrins in tumorigenesis, and both tumor-promoting and suppressive activities have been identified. In this review we discuss the dual role of the laminin-binding integrins α3ß1 and α6ß4 in tumor development and progression, and examine the factors and mechanisms involved in these opposing effects.


Subject(s)
Gene Expression Regulation, Neoplastic , Integrin alpha3beta1/genetics , Integrin alpha6beta4/genetics , Laminin/genetics , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Adhesion , Cell Movement , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Integrin alpha3beta1/metabolism , Integrin alpha6beta4/metabolism , Laminin/metabolism , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Protein Binding , Signal Transduction
20.
J Clin Invest ; 126(11): 4125-4139, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27701149

ABSTRACT

Neutrophils need to penetrate the perivascular basement membrane for successful extravasation into inflamed tissue, but this process is incompletely understood. Recent findings have associated mammalian sterile 20-like kinase 1 (MST1) loss of function with a human primary immunodeficiency disorder, suggesting that MST1 may be involved in immune cell migration. Here, we have shown that MST1 is a critical regulator of neutrophil extravasation during inflammation. Mst1-deficient (Mst1-/-) neutrophils were unable to migrate into inflamed murine cremaster muscle venules, instead persisting between the endothelium and the basement membrane. Mst1-/- neutrophils also failed to extravasate from gastric submucosal vessels in a murine model of Helicobacter pylori infection. Mechanistically, we observed defective translocation of VLA-3, VLA-6, and neutrophil elastase from intracellular vesicles to the surface of Mst1-/- neutrophils, indicating that MST1 is required for this crucial step in neutrophil transmigration. Furthermore, we found that MST1 associates with the Rab27 effector protein synaptotagmin-like protein 1 (JFC1, encoded by Sytl1 in mice), but not Munc13-4, thereby regulating the trafficking of Rab27-positive vesicles to the cellular membrane. Together, these findings highlight a role for MST1 in vesicle trafficking and extravasation in neutrophils, providing an additional mechanistic explanation for the severe immune defect observed in patients with MST1 deficiency.


Subject(s)
Hepatocyte Growth Factor/immunology , Neutrophils/immunology , Proto-Oncogene Proteins/immunology , Secretory Vesicles/immunology , Transendothelial and Transepithelial Migration/immunology , Abdominal Muscles/blood supply , Abdominal Muscles/immunology , Animals , Basement Membrane/immunology , Biological Transport, Active/genetics , Biological Transport, Active/immunology , Gastric Mucosa/chemistry , Gastric Mucosa/immunology , Hepatocyte Growth Factor/genetics , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Integrin alpha3beta1/genetics , Integrin alpha3beta1/immunology , Integrin alpha6beta1/genetics , Integrin alpha6beta1/immunology , Leukocyte Elastase/genetics , Leukocyte Elastase/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Secretory Vesicles/genetics , Transendothelial and Transepithelial Migration/genetics , Venules/immunology , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL