Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 385
Filter
1.
J Extracell Vesicles ; 12(4): e12323, 2023 04.
Article in English | MEDLINE | ID: mdl-37073802

ABSTRACT

Extracellular vesicles (EVs) influence a host of normal and pathophysiological processes in vivo. Compared to soluble mediators, EVs can traffic a wide range of proteins on their surface including extracellular matrix (ECM) binding proteins, and their large size (∼30-150 nm) limits diffusion. We isolated EVs from the MCF10 series-a model human cell line of breast cancer progression-and demonstrated increasing presence of laminin-binding integrins α3ß1 and α6ß1 on the EVs as the malignant potential of the MCF10 cells increased. Transport of the EVs within a microfluidic device under controlled physiological interstitial flow (0.15-0.75 µm/s) demonstrated that convection was the dominant mechanism of transport. Binding of the EVs to the ECM enhanced the spatial concentration and gradient, which was mitigated by blocking integrins α3ß1 and α6ß1. Our studies demonstrate that convection and ECM binding are the dominant mechanisms controlling EV interstitial transport and should be leveraged in nanotherapeutic design.


Subject(s)
Extracellular Vesicles , Laminin , Humans , Laminin/metabolism , Convection , Integrin alpha6beta1/metabolism , Extracellular Vesicles/metabolism , Integrin alpha3beta1/metabolism , Extracellular Matrix/metabolism
2.
ACS Appl Mater Interfaces ; 15(12): 15084-15095, 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-36926803

ABSTRACT

Tissue engineering advancements have made it possible to modify biomaterials to reconstruct a similar three-dimensional structure of the extracellular matrix (ECM) for follicle development and to supply the required biological signals. We postulated that an artificial polysaccharide hydrogel modified with an ECM mimetic peptide may produce efficient irritation signals by binding to specific integrins providing a suitable environment for follicular development and influencing the behavior of human granulosa cells (hGCs). Laminin, an important component of the extracellular matrix, can modulate hGCs and oocyte growth. Specifically, follicles of mice were randomly divided into two-dimensional (2D) and three-dimensional (3D) culture systems established by a hydrogel modified with RGD or laminin mimetic peptides (IKVAV and YIGSR) and RGD (IYR). Our results showed that 3D cultured systems significantly improved follicle survival, growth, and viability. IYR peptides enhanced the oocyte meiosis competence. Additionally, we explored the effect of 3D culture on hGCs, which improved hGCs viability, increased the proportion of S- and G2/M-phase cells, and inhibited cell apoptosis of hGCs. On days 1 and 2, the secretion of progesterone was reduced in 3D-cultured hGCs. Notably, 3D-cultured hGCs exhibited delayed senescence, decreased oxidative stress, and elevated mitochondrial membrane potential. Moreover, the expression levels of cumulus expansion-related genes (COX2, HAS2, and PTX3) and integrin α6ß1 were upregulated in 3D-cultured hGCs. In conclusion, a 3D culture utilizing hydrogels modified with Laminin-mimetic peptides can provide a durable physical environment suitable for follicular development. The laminin-mimetic peptides may regulate the biological activity of hGCs by attaching to the integrin α6ß1.


Subject(s)
Granulosa Cells , Laminin , Female , Humans , Mice , Animals , Laminin/pharmacology , Integrin alpha6beta1/metabolism , Granulosa Cells/metabolism , Hydrogels/pharmacology , Peptides/pharmacology , Peptides/metabolism , Oligopeptides/pharmacology , Oligopeptides/metabolism , Oxidative Stress
3.
Signal Transduct Target Ther ; 7(1): 290, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36042225

ABSTRACT

Hepatic progenitor cells (HPCs) hold tremendous potential for liver regeneration, but their well-known limitation of proliferation hampers their broader use. There is evidence that laminin is required for the proliferation of HPCs, but the laminin isoform that plays the dominant role and the key intracellular downstream targets that mediate the regulation of HPC proliferation have yet to be determined. Here we showed that p53 expression increased gradually and reached maximal levels around 8 days when laminin α4, α5, ß2, ß1, and γ1 subunit levels also reached a maximum during HPC activation and expansion. Laminin-521 (LN-521) promoted greater proliferation of HPCs than do laminin, matrigel or other laminin isoforms. Inactivation of p53 by PFT-α or Ad-p53V143A inhibited the promotion of proliferation by LN-521. Further complementary MRI and bioluminescence imaging analysis showed that p53 inactivation decreased the proliferation of transplanted HPCs in vivo. p53 was activated by LN-521 through the Integrin α6ß1/FAK-Src-Paxillin/Akt axis. Activated p53 was involved in the nuclear translocation of CDK4 and inactivation of Rb by inducing p27Kip1. Taken together, this study identifies LN-521 as an ideal candidate substrate for HPC culture and uncovers an unexpected positive role for p53 in regulating proliferation of HPCs, which makes it a potential target for HPC-based regenerative medicine.


Subject(s)
Laminin , Tumor Suppressor Protein p53 , Cell Proliferation/genetics , Integrin alpha6beta1/metabolism , Laminin/genetics , Laminin/metabolism , Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics
4.
Stem Cells Dev ; 31(21-22): 706-719, 2022 11.
Article in English | MEDLINE | ID: mdl-35726387

ABSTRACT

In human induced pluripotent stem cells (hiPSCs), laminin-511/α6ß1 integrin interacts with E-cadherin, an intercellular adhesion molecule, to induce the activation of the phosphatidylinositol 3-kinase (PI3K)-dependent signaling pathway. The interaction between laminin-511/α6ß1 integrin and E-cadherin, an intercellular adhesion molecule, results in protection against apoptosis through the proto-oncogene tyrosine-protein kinase Fyn(Fyn)-RhoA-ROCK signaling pathway and the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathway for cell death). In this article, the impact of laminin-511 on hiPSC on α6ß1 integrin-Fyn-RhoA-ROCK signaling is discussed and explored along with validation experiments. PIK3CA mRNA (mean [standard deviation {SD}]: iMatrix-511, 1.00 [0.61]; collagen+MFGE8, 0.023 [0.02]; **P < 0.01; n = 6) and PIK3R1 mRNA (mean [SD]: iMatrix-511, 1.00 [0.79]; collagen+MFGE8, 0.040 [0.06]; *P < 0.05; n = 6) were upregulated by iMatrix-511 resulting from an increased expression of Integrin α6 mRNA (mean [SD]: iMatrix-511, 1.00 [0.42]; collagen+MFGE8, 0.23 [0.05]; **P < 0.01; n = 6). The iMatrix-511 increased the expression of p120-Catenin mRNA (mean [SD]: iMatrix-511, 1.00 [0.71]; collagen+MFGE8, 0.025 [0.03]; **P < 0.01; n = 6) and RAC1 mRNA (mean [SD]: iMatrix-511, 1.00 [0.28]; collagen+MFGE8, 0.39 [0.15]; **P < 0.01; n = 6) by increasing the expression of E-cadherin mRNA (mean [SD]: iMatrix-511, 1.00 [0.38]; collagen+MFGE8, 0.16 [0.11]; **P < 0.01; n = 6). As a result, iMatrix-511 increased the expression of P190 RhoGAP (GTPase-activating proteins) mRNA, such as ARHGAP1 mRNA (mean [SD]: iMatrix-511, 1.00 [0.57]; collagen+MFGE8, 0.032 [0.03]; **P < 0.01; n = 6), ARHGAP4 mRNA (mean [SD]: iMatrix-511, 1.00 [0.56]; collagen+MFGE8, 0.039 [0.049]; **P < 0.01; n = 6), and ARHGAP5 mRNA (mean [SD]: iMatrix-511, 1.00 [0.39]; collagen+MFGE8, 0.063 [0.043]; **P < 0.01; n = 6). Western blotting showed that phospho-Rac1 remained in the cytoplasm and phospho-Fyn showed nuclear transition in iPSCs cultured on iMatrix-511. Proteome analysis showed that PI3K signaling was enhanced and cytoskeletal actin was activated in iPSCs cultured on iMatrix-511. In conclusion, laminin-511 strongly activated the cell survival by promoting α6ß1 integrin-Fyn-RhoA-ROCK signaling in hiPSCs.


Subject(s)
Induced Pluripotent Stem Cells , Laminin , Humans , Integrin alpha6beta1/metabolism , Laminin/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , Induced Pluripotent Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Collagen/metabolism , RNA, Messenger , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
5.
Front Immunol ; 13: 810671, 2022.
Article in English | MEDLINE | ID: mdl-35547732

ABSTRACT

Autoimmune hepatitis (AIH) is a chronic inflammatory liver disease with unknown etiology. CCN1, an extracellular matrix-associated protein, is associated with carcinoma, inflammation, liver fibrosis, and even autoimmune diseases. However, the role that CCN1 plays in AIH has remained undetermined. In this study, expression of CCN1 in liver was detected by real-time PCR, western blot and immunohistochemistry (IHC). CCN1 level in serum was detected by ELISA. Diagnostic value of CCN1 was determined by receiver operating characteristic (ROC) curve analysis. CCN1 conditional knockout (CCN1 fl/fl Cre+) mice were generated by mating CCN1 fl/fl C57BL/6J and CAG-Cre-ERT C57BL/6J mice. Autoimmune hepatitis mice model was induced by concanavalin A (ConA). IKKα/ß, IκBα, NF-κB p65 and Akt phosphorylation were determined by western blot. NF-κB p65 nuclear translocation was examined by immunofluorescence. Here, we found that CCN1 was over-expressed in hepatocytes of AIH patients. CCN1 level also increased in serum of AIH patients compared to healthy controls (HC). ROC curve analysis results showed that serum CCN1 was able to distinguish AIH patients from HD. In ConA induced hepatitis mice model, CCN1 conditional knockout (CCN1 fl/fl Cre+) attenuated inflammation by reducing ALT/AST level and IL-6 expression. In vitro, CCN1 treatment dramatically induced IL-6 production in LO2 cells. Moreover, the production of IL-6 was attenuated by CCN1 knockdown. Furthermore, we showed that CCN1 could activate IL-6 production via the PI3K/Akt/NF-κB signaling pathway by binding to α6ß1 receptor. In summary, our results reveal a novel role of CCN1 in promoting inflammation by upregulation of IL-6 production in AIH. Our study also suggests that targeting of CCN1 may represent a novel strategy in AIH treatment.


Subject(s)
Hepatitis, Autoimmune , NF-kappa B , Animals , Concanavalin A , Cysteine-Rich Protein 61 , Disease Models, Animal , Hepatitis, Autoimmune/etiology , Humans , Inflammation/complications , Integrin alpha6beta1 , Interleukin-6 , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt
6.
Aging (Albany NY) ; 14(3): 1200-1213, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35148282

ABSTRACT

CCN1/CYR61 promotes angiogenesis, tumor growth and chemoresistance by binding to its integrin receptor αvß3 in endothelial and breast cancer (BC) cells. CCN1 controls also tissue regeneration by engaging its integrin receptor α6ß1 to induce fibroblast senescence. Here, we explored if the ability of CCN1 to drive an endocrine resistance phenotype in estrogen receptor-positive BC cells relies on interactions with either αvß3 or α6ß1. First, we took advantage of site-specific mutagenesis abolishing the CCN1 receptor-binding sites to αvß3 and α6ß1 to determine the integrin partner responsible for CCN1-driven endocrine resistance. Second, we explored a putative nuclear role of CCN1 in regulating ERα-driven transcriptional responses. Retroviral forced expression of a CCN1 derivative with a single amino acid change (D125A) that abrogates binding to αvß3 partially phenocopied the endocrine resistance phenotype induced upon overexpression of wild-type (WT) CCN1. Forced expression of the CCN1 mutant TM, which abrogates all the T1, H1, and H2 binding sites to α6ß1, failed to bypass the estrogen requirement for anchorage-independent growth or to promote resistance to tamoxifen. Wild-type CCN1 promoted estradiol-independent transcriptional activity of ERα and enhanced ERα agonist response to tamoxifen. The α6ß1-binding-defective TM-CCN1 mutant lost the ERα co-activator-like behavior of WT-CCN1. Co-immunoprecipitation assays revealed a direct interaction between endogenous CCN1 and ERα, and in vitro approaches confirmed the ability of recombinant CCN1 to bind ERα. CCN1 signaling via α6ß1, but not via αvß3, drives an endocrine resistance phenotype that involves a direct binding of CCN1 to ERα to regulate its transcriptional activity in ER+ BC cells.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Breast Neoplasms/genetics , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Estrogen Receptor alpha/genetics , Female , Humans , Integrin alpha6beta1/metabolism , Integrins , Tamoxifen/pharmacology
7.
Respir Res ; 22(1): 265, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34666752

ABSTRACT

RATIONALE: αv integrins, key regulators of transforming growth factor-ß activation and fibrogenesis in in vivo models of pulmonary fibrosis, are expressed on abnormal epithelial cells (αvß6) and fibroblasts (αvß1) in fibrotic lungs. OBJECTIVES: We evaluated multiple αv integrin inhibition strategies to assess which most effectively reduced fibrogenesis in explanted lung tissue from patients with idiopathic pulmonary fibrosis. METHODS: Selective αvß6 and αvß1, dual αvß6/αvß1, and multi-αv integrin inhibitors were characterized for potency, selectivity, and functional activity by ligand binding, cell adhesion, and transforming growth factor-ß cell activation assays. Precision-cut lung slices generated from lung explants from patients with idiopathic pulmonary fibrosis or bleomycin-challenged mouse lungs were treated with integrin inhibitors or standard-of-care drugs (nintedanib or pirfenidone) and analyzed for changes in fibrotic gene expression or TGF-ß signaling. Bleomycin-challenged mice treated with dual αvß6/αvß1 integrin inhibitor, PLN-74809, were assessed for changes in pulmonary collagen deposition and Smad3 phosphorylation. MEASUREMENTS AND MAIN RESULTS: Inhibition of integrins αvß6 and αvß1 was additive in reducing type I collagen gene expression in explanted lung tissue slices from patients with idiopathic pulmonary fibrosis. These data were replicated in fibrotic mouse lung tissue, with no added benefit observed from inhibition of additional αv integrins. Antifibrotic efficacy of dual αvß6/αvß1 integrin inhibitor PLN-74809 was confirmed in vivo, where dose-dependent inhibition of pulmonary Smad3 phosphorylation and collagen deposition was observed. PLN-74809 also, more potently, reduced collagen gene expression in fibrotic human and mouse lung slices than clinically relevant concentrations of nintedanib or pirfenidone. CONCLUSIONS: In the fibrotic lung, dual inhibition of integrins αvß6 and αvß1 offers the optimal approach for blocking fibrogenesis resulting from integrin-mediated activation of transforming growth factor-ß.


Subject(s)
Antifibrotic Agents/pharmacology , Epithelial Cells/drug effects , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Integrin alpha6beta1/antagonists & inhibitors , Lung/drug effects , Receptors, Vitronectin/antagonists & inhibitors , Animals , Bleomycin , Cell Line , Coculture Techniques , Collagen Type I, alpha 1 Chain/genetics , Collagen Type I, alpha 1 Chain/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Integrin alpha6beta1/metabolism , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Phosphorylation , Receptors, Vitronectin/metabolism , Signal Transduction , Smad3 Protein/metabolism
8.
Transfus Apher Sci ; 60(6): 103285, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34620563

ABSTRACT

The most promising therapy for leukemia is hematopoietic stem cell transplantation. Engraftment of HPSCs mainly depends on some factors such as adhesion molecules, including VLAs. This study tried to delineate the relationship between HPSCs engraftment and expression level of PSGL1 and VLA4, 5, and 6 genes in candidate MM patients for autologous bone marrow transplantation. Firstly, the CD 34+ HPSCs were collected from multiple myeloma (MM) patients after five days of G-CSF therapy through apheresis processes. Then, the patients were categorized into two groups of good and bad prognosis depending on engraftment time (Less or more than 18 days). Followingly, the expression of PSGL1 and VLA4, VLA5, and VLA6 genes were assessed by the qRT-PCR technique in each patient. Finally, the correlation between the genes and engraftment time was investigated to determine the prognostic role of each gene on HPSCs transplantation. Our findings demonstrated that there is a significant correlation between VLA4 (P=< 0.0001) and 5 (P = 0.005) levels and HPSCs engraftment time. As the higher levels of VLA4 and 5, the shorter time HPSCs engraftment occurs. In contrast, there was no significant correlation between VLA6 (P = 0.2) and PSGL1 (P = 0.3) genes levels and engraftment time. So that, the patients with a good prognosis had a higher level of VLA4 and VLA5, but no relation was found between VLA6 and PSGL1. It is concluded that VLA4 and VLA5 expression could be considered a significant prognostic factor for HPSC transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Integrin alpha6beta1/metabolism , Membrane Glycoproteins/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Transplantation Conditioning/methods , Transplantation, Autologous/methods , Adult , Aged , Female , Humans , Male , Middle Aged , Multiple Myeloma/pathology
9.
Micron ; 148: 103106, 2021 09.
Article in English | MEDLINE | ID: mdl-34171483

ABSTRACT

Integrin α2ß1 is a widely expressed collagen I receptor which also mediates laminin-111 binding in some cell types, but the functional relevance of collagen versus laminin binding for different cell types is poorly understood. Here we use AFM-based singe-cell force spectroscopy (SCFS) to compare α2ß1-mediated adhesion strength to collagen and laminin in different cell types. Chinese Hamster Ovary (CHO) cells stably expressing integrin α2ß1 (CHO-A2) displayed enhanced adhesion to collagen, but weak adhesion to laminin, consistent with a role of α2ß1 as a receptor only for collagen in these cells. Inversely, the α2ß1-deficient CHO wildtype cells (CHO-WT) showed weak adhesion to collagen, but strong adhesion to laminin-111, in turn suggesting that integrin α2ß1 expression suppresses laminin binding. Analogous results were obtained in a pair of SAOS-2 human osteosarcoma cell lines. Again, wildtype cells (SAOS-WT) adhered strongly to laminin and poorly to collagen, while expression of integrin α2ß1 (SAOS-A2) induced strong adhesion to collagen, but reduced adhesion to laminin. Expression of α2ß1 also shifted cell spreading preference from laminin to collagen and suppressed laminin-dependent transmigration. In agreement with reduced laminin adhesion, α2ß1 expression downregulated transcription and expression of integrin subunits α6 and ß4, components of the main laminin-111 binding receptors integrin α6ß1 and α6ß4 in these cells. Integrin α6 and ß4 expression was also reduced when α2 expression was chemically induced using tetradecanoyl-phorbol-acetate (TPA). Our results thus show that integrin α2ß1 expression negatively regulates integrin α6ß1 and α6ß4-mediated adhesion, spreading and invasion on laminin in different cancer cell types. In contrast to SAOS-WT, but similar to SAOS-A2 osteosarcoma cells, primary Human osteoblasts (HOB) cells express α2 but only low levels of ß4 integrin, preferentially adhere to and spread on collagen over laminin and show suppressed laminin-dependent transmigration. By enhancing collagen binding directly and suppressing laminin binding indirectly through laminin receptor downregulation, α2ß1 expression may thus re-direct migrating cancer cells from laminin-rich to collagenous tissues and partially revert osteosarcoma cells towards an untransformed phenotype.


Subject(s)
Integrin alpha2beta1 , Receptors, Laminin , Animals , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Humans , Integrin alpha2beta1/genetics , Integrin alpha6beta1 , Laminin
10.
Nat Commun ; 12(1): 4012, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34188035

ABSTRACT

Recognition of laminin by integrin receptors is central to the epithelial cell adhesion to basement membrane, but the structural background of this molecular interaction remained elusive. Here, we report the structures of the prototypic laminin receptor α6ß1 integrin alone and in complex with three-chain laminin-511 fragment determined via crystallography and cryo-electron microscopy, respectively. The laminin-integrin interface is made up of several binding sites located on all five subunits, with the laminin γ1 chain C-terminal portion providing focal interaction using two carboxylate anchor points to bridge metal-ion dependent adhesion site of integrin ß1 subunit and Asn189 of integrin α6 subunit. Laminin α5 chain also contributes to the affinity and specificity by making electrostatic interactions with large surface on the ß-propeller domain of α6, part of which comprises an alternatively spliced X1 region. The propeller sheet corresponding to this region shows unusually high mobility, suggesting its unique role in ligand capture.


Subject(s)
Epithelial Cells/metabolism , Integrin alpha6/metabolism , Integrin alpha6beta1/metabolism , Integrin beta1/metabolism , Laminin/metabolism , Amino Acid Sequence , Basement Membrane/metabolism , Binding Sites/physiology , Cell Adhesion/physiology , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Protein Conformation , Protein Domains/physiology , Static Electricity
11.
J Cell Biol ; 219(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-33180092

ABSTRACT

The delivery of glucose and antioxidants is vital to maintain homeostasis and lens transparency. Here, we report a new mechanism whereby mechanically activated connexin (Cx) hemichannels serve as a transport portal for delivering glucose and glutathione (GSH). Integrin α6ß1 in outer cortical lens fiber activated by fluid flow shear stress (FFSS) induced opening of hemichannels. Inhibition of α6 activation prevented hemichannel opening as well as glucose and GSH uptake. The activation of integrin ß1, a heterodimeric partner of α6 in the absence of FFSS, increased Cx50 hemichannel opening. Hemichannel activation by FFSS depended on the interaction of integrin α6 and Cx50 C-terminal domain. Moreover, hemichannels in nuclear fiber were unresponsive owing to Cx50 truncation. Taken together, these results show that mechanically activated α6ß1 integrin in outer cortical lens fibers leads to opening of hemichannels, which transport glucose and GSH into cortical lens fibers. This study unveils a new transport mechanism that maintains metabolic and antioxidative function of the lens.


Subject(s)
Antioxidants/metabolism , Avian Proteins/metabolism , Connexins/metabolism , Glutathione/metabolism , Integrin alpha6beta1/metabolism , Lens, Crystalline/metabolism , Mechanotransduction, Cellular , Animals , Avian Proteins/genetics , Biological Transport, Active , Chick Embryo , Chickens , Connexins/genetics , Integrin alpha6beta1/genetics , Protein Domains
12.
Oncogene ; 39(31): 5390-5404, 2020 07.
Article in English | MEDLINE | ID: mdl-32565538

ABSTRACT

The androgen receptor (AR) is the major driver of prostate cancer growth and survival. However, almost all patients relapse with castration-resistant disease (CRPC) when treated with anti-androgen therapy. In CRPC, AR is often aberrantly activated independent of androgen. Targeting survival pathways downstream of AR could be a viable strategy to overcome CRPC. Surprisingly, little is known about how AR drives prostate cancer survival. Furthermore, CRPC tumors in which Pten is lost are also resistant to eradication by PI3K inhibitors. We sought to identify the mechanism by which AR drives tumor survival in CRPC to identify ways to overcome resistance to PI3K inhibition. We found that integrins α6ß1 and Bnip3 are selectively elevated in CRPC downstream of AR. While integrin α6 promotes survival and is a direct transcriptional target of AR, the ability of AR to induce Bnip3 is dependent on adhesion to laminin and integrin α6ß1-dependent nuclear translocation of HIF1α. Integrins α6ß1 and Bnip3 were found to promote survival of CRPC cells selectively on laminin through the induction of autophagy and mitophagy. Furthermore, blocking Bnip3 or integrin α6ß1 restored sensitivity to PI3K inhibitors in Pten-negative CRPC. We identified an AR driven pathway that cooperates with laminin and hypoxia to drive resistance to PI3K inhibitors. These findings can help explain in part why PI3K inhibitors have failed in clinical trials to overcome AR-dependent CRPC.


Subject(s)
Integrin alpha6beta1/metabolism , Membrane Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Androgen/genetics , Animals , Humans , Male , Protein Kinase Inhibitors/therapeutic use , Receptors, Androgen/metabolism , Survival Analysis
13.
J Exp Med ; 217(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32379272

ABSTRACT

The endothelial cell basement membrane (BM) is a barrier to migrating leukocytes and a rich source of signaling molecules that can influence extravasating cells. Using mice lacking the major endothelial BM components, laminin 411 or 511, in murine experimental autoimmune encephalomyelitis (EAE), we show here that loss of endothelial laminin 511 results in enhanced disease severity due to increased T cell infiltration and altered polarization and pathogenicity of infiltrating T cells. In vitro adhesion and migration assays reveal higher binding to laminin 511 than laminin 411 but faster migration across laminin 411. In vivo and in vitro analyses suggest that integrin α6ß1- and αvß1-mediated binding to laminin 511-high sites not only holds T cells at such sites but also limits their differentiation to pathogenic Th17 cells. This highlights the importance of the interface between the endothelial monolayer and the underlying BM for modulation of immune cell phenotype.


Subject(s)
Basement Membrane/metabolism , Brain/pathology , Endothelial Cells/metabolism , T-Lymphocytes/immunology , Animals , Basement Membrane/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/drug effects , Integrin alpha6beta1/metabolism , Laminin/pharmacology , Mice , Receptors, Vitronectin/metabolism , Spinal Cord/pathology , T-Lymphocytes/drug effects , Th17 Cells/drug effects , Th17 Cells/metabolism
14.
Circulation ; 139(25): 2877-2891, 2019 06 18.
Article in English | MEDLINE | ID: mdl-30917686

ABSTRACT

BACKGROUND: Atherosclerosis occurs preferentially at the blood vessels encountering blood flow turbulence. The matricellular protein CCN1 is induced in endothelial cells by disturbed flow, and is expressed in advanced atherosclerotic lesions in patients and in the Apoe-/- mouse model. The role of CCN1 in atherosclerosis remains undefined. METHODS: To assess the function of CCN1 in vivo, knock-in mice carrying the integrin α6ß1-binding-defective mutant allele Ccn1-dm on the Apoe-/- background were tested in an atherosclerosis model generated by carotid artery ligation. Additionally, CCN1-regulated functional phenotypes of human umbilical vein endothelial cells, or primary mouse aortic endothelial cells isolated from wild-type and Ccn1 dm/dm mice, were investigated in the in vitro shear stress experiments under unidirectional laminar shear stress (12 dyn/cm2) versus oscillatory shear stress (±5 dyn/cm2) conditions. RESULTS: We found that Ccn1 expression was upregulated in the arterial endothelium 3 days after ligation before any detectable structural changes, and intensified with the progression of atherosclerotic lesions. Compared with Apoe-/- controls, Ccn1 dm/dm/ Apoe-/- mice were remarkably resistant to ligation-induced plaque formation (n=6). These mice exhibited lower oxidative stress, expression of endothelin-1 and monocyte chemoattractant protein-1, and monocyte homing. CCN1/α6ß1 critically mediated flow-induced activation of the pleiotropic transcription factor nuclear factor-κB and therefore the induction of atheroprone gene expression in the mouse arterial endothelium after ligation (n=6), or in cultured human umbilical vein endothelial cells or primary mouse aortic endothelial cells exposed to oscillatory shear stress (n=3 in triplicate). Interestingly, the activation of nuclear factor-κB by CCN1/α6ß1 signaling prompted more production of CCN1 and α6ß1. Blocking CCN1-α6ß1 binding by the Ccn1-dm mutation or by T1 peptide (derived from an α6ß1-binding sequence of CCN1) disrupted the positive-feedback regulation between CCN1/α6ß1 and nuclear factor-κB, and prevented flow-induced atheroprone phenotypic alterations in endothelial cells or atherosclerosis in mice. CONCLUSIONS: These data demonstrate a causative role of CCN1 in atherosclerosis via modulating endothelial phenotypes. CCN1 binds to its receptor integrin α6ß1 to activate nuclear factor-κB, thereby instigating a vicious circle to persistently promote atherogenesis. T1, a peptide antagonist selectively targeting CCN1-α6ß1, can be further optimized for developing T1-mimetics to treat atherosclerosis.


Subject(s)
Carotid Artery Diseases/metabolism , Carotid Artery, Common/metabolism , Cysteine-Rich Protein 61/metabolism , Endothelial Cells/metabolism , Mechanotransduction, Cellular , Plaque, Atherosclerotic , Animals , Carotid Artery Diseases/diagnosis , Carotid Artery Diseases/pathology , Carotid Artery Diseases/physiopathology , Carotid Artery, Common/pathology , Carotid Artery, Common/physiopathology , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Integrin alpha6beta1/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mutation , NF-kappa B/metabolism , Phenotype , Regional Blood Flow , Stress, Mechanical
15.
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
16.
J Cell Physiol ; 234(6): 9297-9307, 2019 06.
Article in English | MEDLINE | ID: mdl-30317661

ABSTRACT

In recent years, osteosarcoma survival rates have failed to improve significantly with conventional treatment modalities because of the development of chemotherapeutic resistance. The human breast cancer resistance protein/ATP binding cassette subfamily G member 2 (BCRP/ABCG2), a member of the ATP-binding cassette family, uses ATP hydrolysis to expel xenobiotics and chemotherapeutics from cells. CCN family member 2 (CCN2) is a secreted protein that modulates the biological function of cancer cells, enhanced ABCG2 protein expression and activation in this study via the α6ß1 integrin receptor and increased osteosarcoma cell viability. CCN2 treatment downregulated miR-519d expression, which promoted ABCG2 expression. In a mouse xenograft model, knockdown of CCN2 expression increased the therapeutic effect of doxorubicin, which was reversed by ABCG2 overexpression. Our data show that CCN2 increases ABCG2 expression and promotes drug resistance through the α6ß1 integrin receptor, whereas CCN2 downregulates miR-519d. CCN2 inhibition may represent a new therapeutic concept in osteosarcoma.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Connective Tissue Growth Factor/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Osteosarcoma/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Survival/genetics , Humans , Integrin alpha6beta1/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Signal Transduction
17.
J Biol Chem ; 293(41): 16006-16018, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30150299

ABSTRACT

Group A Streptococcus (GAS) is a human-specific pathogen responsible for a wide range of diseases, ranging from superficial to life-threatening invasive infections, including endometritis, and autoimmune sequelae. GAS strains express a vast repertoire of virulence factors that varies depending on the strain genotype, and many adhesin proteins that enable GAS to adhere to host cells are restricted to some genotypes. GAS emm28 is the third most prevalent genotype in invasive infections in France and is associated with gyneco-obstetrical infections. emm28 strains harbor R28, a cell wall-anchored surface protein that has previously been reported to promote adhesion to cervical epithelial cells. Here, using cellular and biochemical approaches, we sought to determine whether R28 supports adhesion also to other cells and to characterize its cognate receptor. We show that through its N-terminal domain, R28Nt, R28 promotes bacterial adhesion to both endometrial-epithelial and endometrial-stromal cells. R28Nt was further subdivided into two domains, and we found that both are involved in cell binding. R28Nt and both subdomains interacted directly with the laminin-binding α3ß1, α6ß1, and α6ß4 integrins; interestingly, these bindings events did not require divalent cations. R28 is the first GAS adhesin reported to bind directly to integrins that are expressed in most epithelial cells. Finally, R28Nt also promoted binding to keratinocytes and pulmonary epithelial cells, suggesting that it may be involved in supporting the prevalence in invasive infections of the emm28 genotype.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Adhesion/physiology , Integrin alpha3beta1/metabolism , Integrin alpha6beta1/metabolism , Integrin alpha6beta4/metabolism , Adhesins, Bacterial/chemistry , Bacterial Adhesion/physiology , Bacterial Proteins/chemistry , Cell Line, Tumor , Endometrium/metabolism , Epithelial Cells/metabolism , Female , Humans , Keratinocytes/metabolism , Protein Binding , Protein Domains , Streptococcus pyogenes/chemistry , Stromal Cells/metabolism
18.
Sci Transl Med ; 10(453)2018 08 08.
Article in English | MEDLINE | ID: mdl-30089633

ABSTRACT

Autoimmune pancreatitis (AIP), a major manifestation of immunoglobulin G4-related disease (IgG4-RD), is an immune-mediated disorder, but the target autoantigens are still unknown. We previously reported that IgG in patients with AIP induces pancreatic injuries in mice by binding the extracellular matrix (ECM). In the current study, we identified an autoantibody against laminin 511-E8, a truncated laminin 511, one of the ECM proteins, in patients with AIP. Anti-laminin 511-E8 IgG was present in 26 of 51 AIP patients (51.0%), but only in 2 of 122 controls (1.6%), by enzyme-linked immunosorbent assay. Because truncated forms of other laminin family members in other organs have been reported, we confirmed that truncated forms of laminin 511 also exist in human and mouse pancreas. Histologic studies with patient pancreatic tissues showed colocalization of patient IgG and laminin 511. Immunization of mice with human laminin 511-E8 induced antibodies and pancreatic injury, fulfilling the pathologic criteria for human AIP. Four of 25 AIP patients without laminin 511-E8 antibodies had antibodies against integrin α6ß1, a laminin 511 ligand. AIP patients with laminin 511-E8 antibodies exhibited distinctive clinical features, as the frequencies of malignancies or allergic diseases were significantly lower in patients with laminin 511-E8 antibodies than in those without. The discovery of these autoantibodies should aid in the understanding of AIP pathophysiology and possibly improve the diagnosis of AIP.


Subject(s)
Antigens/blood , Autoimmune Diseases/blood , Laminin/blood , Pancreatitis/blood , Aged , Animals , Autoantibodies/blood , Autoimmune Diseases/immunology , Female , Humans , Immunization , Immunoglobulin G/blood , Integrin alpha6beta1/metabolism , Male , Mice , Mice, Inbred BALB C , Middle Aged , Pancreatitis/immunology
19.
Brain Res ; 1700: 78-85, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30006296

ABSTRACT

The laminin family of glycoproteins are major constituents of the basal lamina of blood vessels, and play a fundamental role in promoting endothelial differentiation and blood-brain barrier (BBB) stability. Chronic mild hypoxia (CMH), in which mice are exposed to 8% O2 for two weeks, induces a strong vascular remodeling response in the central nervous system (CNS) that includes endothelial proliferation, angiogenesis, arteriogenesis as well as increased expression of tight junction proteins, suggestive of enhanced vascular integrity. As previous studies highlight an important role for laminin in promoting vascular differentiation and BBB stability, the goal of this study was to determine if CMH influences the expression of the laminins and their cell surface receptors in cerebral blood vessels. Our studies revealed that over a 14 day period of CMH, blood vessels in the brain showed strong upregulation of the specific laminin subunits α1 and α4, corresponding to increased expression of laminins 111 and 411 respectively, with no discernible changes in the expression levels of the α2 or α5 laminin subunits. This was accompanied by marked endothelial upregulation of the laminin receptor α6ß1 integrin but no alterations in the other laminin receptors α1ß1 integrin or dystroglycan. In light of the instructive role for laminins in promoting vascular differentiation and stability, these data suggest that upregulation of the laminin-α6ß1 integrin axis is part of the molecular response triggered by mild hypoxia that leads to enhanced BBB stability.


Subject(s)
Blood-Brain Barrier/metabolism , Hypoxia/metabolism , Integrin alpha6beta1/metabolism , Laminin/metabolism , Animals , Blood-Brain Barrier/pathology , Cerebrovascular Disorders/metabolism , Cerebrovascular Disorders/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Hypoxia/pathology , Mice , Vascular Remodeling/physiology
20.
J Invest Dermatol ; 138(8): 1825-1833, 2018 08.
Article in English | MEDLINE | ID: mdl-29510193

ABSTRACT

Fibroblast-derived melanogenic paracrine mediators are known to play a role in melanogenesis. To investigate the effect of CCN1 (also called CYR61 or cysteine-rich 61) on melanogenesis, normal human epidermal melanocytes were treated with recombinant CCN1 protein. Our findings show that CCN1 activates melanogenesis through promoting melanosome maturation and up-regulation of MITF, TRP-1, and tyrosinase via the integrin α6ß1, p38 MAPK, and ERK signaling pathways. Furthermore, we found that UVB irradiation stimulates the secretion of CCN1 from normal human dermal fibroblasts, and CCN1 knockdown in fibroblasts attenuates melanogenesis in melanocyte-fibroblast co-culture system. Moreover, using immunohistochemistry and immunofluorescence staining, we discovered that CCN1 is overexpressed in the dermis of both solar lentigines and Riehl's melanosis lesions. These findings suggest that CCN1 is a fibroblast-derived melanogenic paracrine mediator that is secreted under UVB irradiation, and it may play an important role in the development of hyperpigmentation diseases such as Riehl's melanosis.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Fibroblasts/metabolism , Lentigo/pathology , MAP Kinase Signaling System , Melanins/biosynthesis , Melanocytes/metabolism , Adult , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Dermis/pathology , Dermis/radiation effects , Epidermis/metabolism , Female , Fibroblasts/radiation effects , Gene Knockdown Techniques , Humans , Integrin alpha6beta1/metabolism , Lentigo/etiology , Male , Middle Aged , Ultraviolet Rays , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...