Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 208
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33903257

ABSTRACT

The role of integrins, in particular αv integrins, in regulating insulin resistance is incompletely understood. We have previously shown that the αvß5 integrin ligand milk fat globule epidermal growth factor like 8 (MFGE8) regulates cellular uptake of fatty acids. In this work, we evaluated the impact of MFGE8 on glucose homeostasis. We show that acute blockade of the MFGE8/ß5 pathway enhances while acute augmentation dampens insulin-stimulated glucose uptake. Moreover, we find that insulin itself induces cell-surface enrichment of MFGE8 in skeletal muscle, which then promotes interaction between the αvß5 integrin and the insulin receptor leading to dampening of skeletal-muscle insulin receptor signaling. Blockade of the MFGE8/ß5 pathway also enhances hepatic insulin sensitivity. Our work identifies an autoregulatory mechanism by which insulin-stimulated signaling through its cognate receptor is terminated through up-regulation of MFGE8 and its consequent interaction with the αvß5 integrin, thereby establishing a pathway that can potentially be targeted to improve insulin sensitivity.


Subject(s)
Antigens, Surface/genetics , Insulin Resistance/genetics , Insulin/genetics , Milk Proteins/genetics , Receptors, Vitronectin/genetics , Animals , Antigens, CD/genetics , Fatty Acids/genetics , Fatty Acids/metabolism , Glucose/metabolism , Glycolipids/genetics , Glycoproteins/genetics , Homeostasis/genetics , Humans , Integrin alphaVbeta3/genetics , Lipid Droplets , Mice , Muscle, Skeletal/metabolism , Receptor, Insulin/genetics , Signal Transduction/genetics
2.
FASEB J ; 36(7): e22389, 2022 07.
Article in English | MEDLINE | ID: mdl-35657599

ABSTRACT

The arginine-glycine-aspartic acid (RGD) motif is a cell adhesion sequence that binds to integrins. Some RGD-containing peptides promote adhesion of both embryonic stem cells and induced pluripotent stem cells (iPSCs); however, not all such RGD-containing peptides are active. In this study, we elucidated the role of RGD-neighboring sequences on iPSC adhesion using diverse synthetic peptides and recombinant proteins. Our results indicate that iPSC adhesion requires RGDX1 X2  sequences, such as RGDVF and RGDNY, and that the X1 X2 residues are essential for the adhesion via integrin αvß5 but not αvß3. iPSCs express integrin αvß5 but not αvß3; therefore, iPSC adhesion requires the RGDX1 X2 -containing sequences. The importance of the X1 X2 residues was confirmed with both HeLa and A549 cells, which express integrin αvß5 but not αvß3. Analysis of RGD-neighboring sequences provides important insights into ligand-binding specificity of integrins. Identification of integrin αvß5-binding motifs is potentially useful in drug development, drug delivery, cell culture, and tissue engineering.


Subject(s)
Pluripotent Stem Cells , Receptors, Vitronectin , Cell Adhesion/physiology , Integrin alphaVbeta3/genetics , Oligopeptides/chemistry , Peptides/chemistry , Pluripotent Stem Cells/metabolism , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism
3.
FASEB J ; 35(4): e21443, 2021 04.
Article in English | MEDLINE | ID: mdl-33749877

ABSTRACT

The effective clearance of apoptotic cells is an essential step in the resolution of healing wounds. In particular, blood vessel regression during wound resolution produces a significant number of apoptotic endothelial cells (ApoEC) that must be cleared. In considering the fate of ApoEC and the presence of fibroblasts during wound resolution, we hypothesized that fibroblasts might serve as phagocytes involved in endothelial cell removal. The current study investigated whether dermal fibroblasts engulf ApoEC, whether this uptake alters the phenotype of dermal fibroblasts, and the biological molecules involved. In both in vitro and in vivo studies, following ApoEC engulfment, fibroblasts acquired a pro-healing phenotype (increased cell migration, contractility, α-smooth muscle actin expression, and collagen deposition). In addition, fibroblast uptake of ApoEC was shown to be mediated in part by the milk fat globule-EGF factor 8 protein/integrin αv ß5 pathway. Our study demonstrates a novel function of fibroblasts in the clearance of ApoEC and suggests that this capability has significant implications for tissue repair and fibrosis.


Subject(s)
Endothelial Cells/metabolism , Skin/blood supply , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Apoptosis , Female , Green Fluorescent Proteins , Humans , Mice, Inbred C57BL , Milk Proteins/genetics , Milk Proteins/metabolism , Phagocytosis , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Wound Healing
4.
J Cell Sci ; 132(20)2019 10 16.
Article in English | MEDLINE | ID: mdl-31515275

ABSTRACT

Diurnal clearance phagocytosis by the retinal pigment epithelium (RPE) is a conserved efferocytosis process whose binding step is mediated by αvß5 integrin receptors. Two related annexins, A5 (ANXA5) and A6 (ANXA6), share an αvß5 integrin-binding motif. Here, we report that ANXA5, but not ANXA6, regulates the binding capacity for spent photoreceptor outer segment fragments or apoptotic cells by fibroblasts and RPE. Similar to αvß5-deficient RPE, ANXA5-/- RPE in vivo lacks the diurnal burst of phagocytosis that follows photoreceptor shedding in wild-type retina. Increasing ANXA5 in cells lacking αvß5 or increasing αvß5 in cells lacking ANXA5 does not affect particle binding. Association of cytosolic ANXA5 and αvß5 integrin in RPE in culture and in vivo further supports their functional interdependence. Silencing ANXA5 is sufficient to reduce levels of αvß5 receptors at the apical phagocytic surface of RPE cells. The effect of ANXA5 on surface αvß5 and on particle binding requires the C-terminal ANXA5 annexin repeat but not its unique N-terminus. These results identify a novel role for ANXA5 specifically in the recognition and binding step of clearance phagocytosis, which is essential to retinal physiology.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Annexin A5/metabolism , Apoptosis , Phagocytosis , Photoreceptor Cells, Vertebrate/metabolism , Receptors, Vitronectin/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Annexin A5/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Photoreceptor Cells, Vertebrate/cytology , Rats , Rats, Sprague-Dawley , Receptors, Vitronectin/genetics , Retinal Pigment Epithelium/cytology
5.
Eur J Immunol ; 48(7): 1181-1187, 2018 07.
Article in English | MEDLINE | ID: mdl-29676784

ABSTRACT

Macrophages play a crucial role in maintaining homeostasis in the intestine, but the underlying mechanisms have not yet been elucidated fully. Here, we show for the first time that mature intestinal macrophages in mouse intestine express high levels of αvß5 integrin, which acts as a receptor for the uptake of apoptotic cells and can activate molecules involved in several aspects of tissue homeostasis such as angiogenesis and remodeling of the ECM. αvß5 is not expressed by other immune cells in the intestine, is already present on intestinal macrophages soon after birth, and its expression is not dependent on the microbiota. In adults, αvß5 is induced during the differentiation of monocytes in response to the local environment and it confers intestinal macrophages with the ability to promote engulfment of apoptotic cells via engagement of the bridging molecule milk fat globule EGF-like molecule 8. In the absence of αvß5, there are fewer monocytes in the mucosa and mature intestinal macrophages have decreased expression of metalloproteases and IL 10. Mice lacking αvß5 on haematopoietic cells show increased susceptibility to chemical colitis and we conclude that αvß5 contributes to the tissue repair by regulating the homeostatic properties of intestinal macrophages.


Subject(s)
Colitis/immunology , Integrin alpha5/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Macrophages/metabolism , Animals , Cells, Cultured , Colitis/chemically induced , Epidermal Growth Factor/metabolism , Gene Expression Regulation , Homeostasis , Humans , Integrin alpha5/genetics , Macrophages/immunology , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Transplantation Chimera
6.
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
7.
Bioconjug Chem ; 30(11): 2777-2781, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31657906

ABSTRACT

Learning from the design concept of antibody-drug conjugates (ADCs), we attempted to construct siRNA conjugated polymer brush by attaching a multiple of siRNA to the units of poly(amino acids) [poly(lysine) derivatives] through an intracellular cleavable disulfide bond. Note that the disulfide linkage is stable at extracellular milieu yet subjected to cleavage into free thiol residues at the intracellular reducing compartments. Consequently, ready release of arrays of active siRNA was achieved selectively in the intracellular compartments. Furthermore, tumor-targeted cyclic Asp-Gly-Arg (RGD) was conjugated to the aforementioned polymer brush in view that the RGD receptors (αVß3 and αVß5 integrins) were overexpressed over a wide spectrum of cancerous cells. Our subsequent results have achieved potent gene silencing in cultured cancerous cells from our proposed siRNA delivery construct. To our best knowledge, our proposed conjugate should be the first example of using an ADC platform in successful intracellular transportation of larger macromolecular biological payloads rather than small molecular chemotherapeutic drugs. Hence, the proposed strategy may serve as a promising avenue for targeted delivery of macromolecular pharmaceutical payloads.


Subject(s)
Gene Silencing , Glioma/genetics , Integrin alphaVbeta3/antagonists & inhibitors , Oligopeptides/chemistry , Polymers/chemistry , RNA, Small Interfering/administration & dosage , Receptors, Vitronectin/antagonists & inhibitors , Drug Delivery Systems , Glioma/metabolism , Glioma/pathology , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Oxidation-Reduction , RNA, Small Interfering/genetics , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Tumor Cells, Cultured
8.
BMC Biol ; 16(1): 92, 2018 08 17.
Article in English | MEDLINE | ID: mdl-30119679

ABSTRACT

BACKGROUND: Increased activity of the receptor tyrosine kinase Tie2 has been implicated in the promotion of pathological angiogenesis. This activity is mainly mediated through angiopoietin (Ang)1- and Ang2-dependent activation of integrins by Tie2, rendering the Ang/Tie2/integrin axis an attractive putative target for cancer therapeutics. RESULTS: To target this axis, we developed single domain, non-immunoglobulin high-affinity bi-specific protein inhibitors against both Tie2 and αvß3 integrin. We have previously engineered the Ang2-binding domain of Tie2 (Ang2-BD) as a Tie2 inhibitor. Here, we engineered an exposed loop in Ang2-BD to generate variants that include an integrin-binding Arg-Gly-Asp (RGD) motif and used flow cytometry screening of a yeast-displayed Ang2-BD RGD loop library to identify the integrin antagonists. The bi-specific antagonists targeting both Tie2 and αvß3 integrin inhibited adhesion and proliferation of endothelial cells cultured together with the αvß3 integrin ligand vitronectin, as well as endothelial cell invasion and tube formation. The bi-specific reagents inhibited downstream signaling by Tie2 intracellularly in response to its agonist Ang1 more effectively than the wild-type Ang2 BD that binds Tie2 alone. CONCLUSIONS: Collectively, this study-the first to describe inhibitors targeting all the known functions resulting from Tie2/integrin αvß3 cross-talk-has created new tools for studying Tie2- and integrin αvß3-dependent molecular pathways and provides the basis for the rational and combinatorial engineering of ligand-Tie2 and ligand-integrin αvß3 receptor interactions. Given the roles of these pathways in cancer angiogenesis and metastasis, this proof of principle study paves the route to create novel Tie2/integrin αvß3-targeting proteins for clinical use as imaging and therapeutic agents.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neovascularization, Physiologic/genetics , Receptor, TIE-2/antagonists & inhibitors , Receptors, Vitronectin/genetics , Ribonuclease, Pancreatic/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Animals , Mice , Receptor, TIE-2/chemistry , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Receptors, Vitronectin/chemistry , Receptors, Vitronectin/metabolism , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism
9.
Angiogenesis ; 21(2): 251-266, 2018 05.
Article in English | MEDLINE | ID: mdl-29299782

ABSTRACT

Spinal cord injury (SCI) leads to rapid destruction of neuronal tissue, resulting in devastating motor and sensory deficits. This is exacerbated by damage to spinal cord blood vessels and loss of vascular integrity. Thus, approaches that protect existing blood vessels or stimulate the growth of new blood vessels might present a novel approach to minimize loss or promote regeneration of spinal cord tissue following SCI. In light of the remarkable power of chronic mild hypoxia (CMH) to stimulate vascular remodeling in the brain, the goal of this study was to examine how CMH (8% O2 for up to 7 days) affects blood vessel remodeling in the spinal cord. We found that CMH promoted the following: (1) endothelial proliferation and increased vascularity as a result of angiogenesis and arteriogenesis, (2) increased vascular expression of the angiogenic extracellular matrix protein fibronectin as well as concomitant increases in endothelial expression of the fibronectin receptor α5ß1 integrin, (3) strongly upregulated endothelial expression of the tight junction proteins claudin-5, ZO-1 and occludin and (4) astrocyte activation. Of note, the vascular remodeling changes induced by CMH were more extensive in white matter. Interestingly, hypoxic-induced vascular remodeling in spinal cord blood vessels was markedly attenuated in mice lacking endothelial α5 integrin expression (α5-EC-KO mice). Taken together, these studies demonstrate the considerable remodeling potential of spinal cord blood vessels and highlight an important angiogenic role for the α5ß1 integrin in promoting endothelial proliferation. They also imply that stimulation of the α5ß1 integrin or controlled use of mild hypoxia might provide new approaches for promoting angiogenesis and improving vascular integrity in spinal cord blood vessels.


Subject(s)
Hypoxia/metabolism , Receptors, Vitronectin/metabolism , Spinal Cord Injuries/metabolism , Vascular Remodeling , White Matter/blood supply , White Matter/metabolism , Animals , Chronic Disease , Hypoxia/genetics , Hypoxia/pathology , Hypoxia/physiopathology , Mice , Mice, Knockout , Receptors, Vitronectin/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , White Matter/pathology , White Matter/physiopathology
10.
J Transl Med ; 16(1): 352, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30541573

ABSTRACT

BACKGROUND: To date the TGF-ß1 activation mediated by integrin ανß5 during fibrosis is well-known. This process has been shown also in the heart, where cardiac fibroblasts (CF) differentiate into α-smooth muscle actin (α-SMA)-positive myofibroblasts (MyoFB). Here, we studied the effects on CF, isolated by spontaneously hypertensive rats (SHR), of integrin ανß5 inhibition in MyoFB differentiation. METHODS: Staining and immunohistochemistry were performed on rat cardiac tissue. CF were isolated by enzymatic digestion from SHR (SHR-CF) and normotensive WKY (WKY-CF) rat hearts and then treated for in vitro evaluation. RESULTS: SHR heart tissues revealed a higher TGF-ß1 expression vs. WKY samples. SHR-CF showed an enhanced SMAD2/3 activation and an up-regulated expression of α-SMA, a typical MyoFB marker, especially after TGF-ß1 treatment. Immunostaining on cardiac tissues revealed a higher expression of integrin ανß5 in SHR vs. WKY rat hearts. In vitro results confirmed the up-regulation of integrin ανß5 expression in SHR-CF at basal condition and after TGF-ß1 treatment, in comparison with WKY-CF. Inhibition of integrin ανß5 by cilengitide treatment led a decreased expression of ανß5, collagen I, and α-SMA in SHR-CF vs. WKY-CF, resulting in a diminished differentiation of CF into MyoFB. Taking together, results suggested that SHR-CF are more susceptible to TGF-ß1, showing an up-regulated activation of SMAD2/3 signaling, and an increased ανß5, α-SMA, and collagen I expression. Hypertension stimulus promoted an up-regulation of integrin ανß5 on SHR cardiac tissue and its in vitro inhibition reverted pro-fibrotic events of SHR-CF. CONCLUSION: Inhibition of integrin ανß5 exerted by cilengitide strongly diminished SHR-CF differentiation into detrimental MyoFB. So, integrin ανß5 might be considered a novel therapeutic target and cilengitide an effective pharmacological tool to limit the progression of hypertension-induced cardiac fibrosis.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , Myocardium/metabolism , Myocardium/pathology , Receptors, Vitronectin/antagonists & inhibitors , Actins/metabolism , Animals , Biomarkers/metabolism , Blood Pressure/drug effects , Collagen Type I/metabolism , Diastole/drug effects , Male , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Snake Venoms/pharmacology , Systole/drug effects , Transforming Growth Factor beta1/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
11.
J Biol Chem ; 290(1): 259-71, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25398877

ABSTRACT

Integrins are activated by signaling from inside the cell (inside-out signaling) through global conformational changes of integrins. We recently discovered that fractalkine activates integrins in the absence of CX3CR1 through the direct binding of fractalkine to a ligand-binding site in the integrin headpiece (site 2) that is distinct from the classical RGD-binding site (site 1). We propose that fractalkine binding to the newly identified site 2 induces activation of site 1 though conformational changes (in an allosteric mechanism). We reasoned that site 2-mediated activation of integrins is not limited to fractalkine. Human secreted phospholipase A2 type IIA (sPLA2-IIA), a proinflammatory protein, binds to integrins αvß3 and α4ß1 (site 1), and this interaction initiates a signaling pathway that leads to cell proliferation and inflammation. Human sPLA2-IIA does not bind to M-type receptor very well. Here we describe that sPLA2-IIA directly activated purified soluble integrin αvß3 and transmembrane αvß3 on the cell surface. This activation did not require catalytic activity or M-type receptor. Docking simulation predicted that sPLA2-IIA binds to site 2 in the closed-headpiece of αvß3. A peptide from site 2 of integrin ß1 specifically bound to sPLA2-IIA and suppressed sPLA2-IIA-induced integrin activation. This suggests that sPLA2-IIA activates αvß3 through binding to site 2. sPLA2-IIA also activated integrins α4ß1 and α5ß1 in a site 2-mediated manner. We recently identified small compounds that bind to sPLA2-IIA and suppress integrin-sPLA2-IIA interaction (e.g. compound 21 (Cmpd21)). Cmpd21 effectively suppressed sPLA2-IIA-induced integrin activation. These results define a novel mechanism of proinflammatory action of sPLA2-IIA through integrin activation.


Subject(s)
Group II Phospholipases A2/chemistry , Integrin alpha4beta1/chemistry , Integrin alphaVbeta3/chemistry , Receptors, Vitronectin/chemistry , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetulus , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Group II Phospholipases A2/antagonists & inhibitors , Group II Phospholipases A2/genetics , Group II Phospholipases A2/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , K562 Cells , Models, Molecular , Molecular Docking Simulation , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Signal Transduction
12.
J Biol Chem ; 289(3): 1467-77, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24293364

ABSTRACT

Tropoelastin protein monomers assemble to form elastin. Cellular integrin αVß3 binds RKRK at the C-terminal tail of tropoelastin. We probed cell interactions with tropoelastin by deleting the RKRK sequence to identify other cell-binding interactions within tropoelastin. We found a novel human dermal fibroblast attachment and spreading site on tropoelastin that is located centrally in the molecule. Inhibition studies demonstrated that this cell adhesion was not mediated by either elastin-binding protein or glycosaminoglycans. Cell interactions were divalent cation-dependent, indicating integrin dependence. Function-blocking monoclonal antibodies revealed that αV integrin(s) and integrin αVß5 specifically were critical for cell adhesion to this part of tropoelastin. These data reveal a common αV integrin-binding theme for tropoelastin: αVß3 at the C terminus and αVß5 at the central region of tropoelastin. Each αV region contributes to fibroblast attachment and spreading, but they differ in their effects on cytoskeletal assembly.


Subject(s)
Fibroblasts/metabolism , Receptors, Vitronectin/metabolism , Tropoelastin/metabolism , Cell Adhesion/physiology , Cell Line , Fibroblasts/cytology , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , Receptors, Vitronectin/genetics , Tropoelastin/genetics
13.
J Biol Chem ; 289(25): 17699-708, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24808173

ABSTRACT

Tenascin-C is an adhesion modulatory matrix protein that is highly expressed in tumors; however, its biochemical activity involved in tumorigenesis is not fully understood. On the other hand, increasing evidence indicates the importance of integrin α5ß1 in cancer development. We previously demonstrated that tenascin-C harbors a functional site that can be released as a proadhesive peptide such as TNIIIA2. Peptide TNIIIA2 is capable of inducing activation of ß1-integrins including α5ß1 via syndecan-4. In this study the proadhesive effect of TNIIIA2 was characterized by potentiated and sustained activation of integrin α5ß1. Based on this effect, TNIIIA2 rendered nontransformed fibroblasts (NIH3T3) resistant to serum deprivation-elicited anoikis through activation of the Akt/Bcl-2 pathway. Moreover, TNIIIA2 hyperstimulated PDGF-dependent proliferation of NIH3T3 by activating integrin α5ß1. Tenascin-C, a parental protein of TNIIIA2, also stimulated PDGF-dependent proliferation, which was blocked by a matrix metalloproteinase-2/9 inhibitor and an anti-TNIIIA2 function-blocking antibody, suggesting proteolytic exposure of the proadhesive effect of TNIIIA2. Mechanistic analyses revealed that TNIIIA2 induced a lateral association of PDGF receptor ß with the molecular complex of activated integrin α5ß1 and syndecan-4 in the membrane microdomains enriched with cholesterol/caveolin-1, resulting in prolonged activation of PDGF receptor ß and the subsequent Ras/mitogen-activated protein kinase pathway in a PDGF-dependent manner. Of note, TNIIIA2 induced continuous proliferation in NIH3T3 in an integrin α5ß1-dependent manner even after they formed a confluent monolayer. Thus, it was proposed that tenascin-C might be involved in deregulated cell growth through potentiated and sustained activation of integrin α5ß1 after exposure of the proadhesive effect of TNIIIA2.


Subject(s)
Cell Proliferation/drug effects , Peptides/pharmacology , Platelet-Derived Growth Factor/metabolism , Receptors, Vitronectin/metabolism , Tenascin/pharmacology , Animals , Cell Survival/drug effects , Humans , K562 Cells , Membrane Microdomains/genetics , Membrane Microdomains/metabolism , Mice , NIH 3T3 Cells , Peptides/chemistry , Platelet-Derived Growth Factor/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Vitronectin/genetics , Syndecan-4/genetics , Syndecan-4/metabolism , Tenascin/chemistry
14.
J Biol Chem ; 289(33): 23141-23153, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24936068

ABSTRACT

The regulation of integrin-mediated adhesion is of vital importance to adaptive and innate immunity. Integrins are versatile proteins and mediate T cell migration and trafficking by binding to extracellular matrix or other cells as well as initiating intracellular signaling cascades promoting survival or activation. The MAPK pathway is known to be downstream from integrins and to regulate survival, differentiation, and motility. However, secondary roles for canonical MAPK pathway members are being discovered. We show that chemical inhibition of RAF by sorafenib or shRNA-mediated knockdown of B-Raf reduces T cell resistance to shear stress to α4ß1 integrin ligands vascular cell adhesion molecule 1 (VCAM-1) and fibronectin, whereas inhibition of MEK/ERK by U0126 had no effect. Microscopy showed that RAF inhibition leads to significant inhibition of T cell spreading on VCAM-1. The association of α4ß1 integrin with the actin cytoskeleton was shown to be dependent on B-Raf activity or expression, whereas α4ß1 integrin affinity for soluble VCAM-1 was not. These effects were shown to be specific for α4ß1 integrin and not other integrins, such as α5ß1 or LFA-1, or a variety of membrane proteins. We demonstrate a novel role for B-Raf in the selective regulation of α4ß1 integrin-mediated adhesion.


Subject(s)
Cytoskeleton/metabolism , Integrin alpha4beta1/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Stress, Physiological/physiology , T-Lymphocytes/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cytoskeleton/genetics , Gene Knockdown Techniques , Humans , Integrin alpha4beta1/genetics , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Shear Strength/drug effects , Shear Strength/physiology , Sorafenib , Stress, Physiological/drug effects , T-Lymphocytes/cytology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
15.
J Cell Biochem ; 116(10): 2385-96, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25857463

ABSTRACT

Colorectal cancer (CRC) metastasis dissemination to secondary sites represents the critical point for the patient's survival. The microenvironment is crucial to cancer progression, influencing tumour cell behaviour by modulating the expression and activation of molecules such as integrins, the cell-extracellular matrix interacting proteins participating in different steps of the tumour metastatic process. In this work, we investigated the role of α5ß1 integrin and how the microenvironment influences this adhesion molecule, in a model of colon cancer progression to the liver. The culture medium conditioned by the IHH hepatic cell line, and the extracellular matrix (ECM) proteins, modulate the activation of α5ß1 integrin in the colon cancer cell line HCT-116, and drives FAK phosphorylation during the process of cell adhesion to fibronectin, one of the main components of liver ECM. In these conditions, α5ß1 modulates the expression/activity of another integrin, α2ß1, involved in the cell adhesion to collagen I. These results suggest that α5ß1 integrin holds a leading role in HCT-116 colorectal cancer cells adhesion to the ECM through the modulation of the intracellular focal adhesion kinase FAK and the α2ß1 integrin activity. The driving role of the tumour microenvironment on CRC dissemination, here detected, and described, strengthens and adds new value to the concept that α5ß1 integrin can be an appropriate and relevant therapeutic target for the control of CRC metastases.


Subject(s)
Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Receptors, Vitronectin/biosynthesis , Tumor Microenvironment/genetics , Cell Line, Tumor , Colorectal Neoplasms/pathology , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Fibronectins/metabolism , Focal Adhesion Kinase 1/biosynthesis , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Neoplasm Metastasis , Phosphorylation , Receptors, Vitronectin/genetics
16.
Nat Mater ; 13(6): 631-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24793358

ABSTRACT

Tissue rigidity regulates processes in development, cancer and wound healing. However, how cells detect rigidity, and thereby modulate their behaviour, remains unknown. Here, we show that sensing and adaptation to matrix rigidity in breast myoepithelial cells is determined by the bond dynamics of different integrin types. Cell binding to fibronectin through either α5ß1 integrins (constitutively expressed) or αvß6 integrins (selectively expressed in cancer and development) adapts force generation, actin flow and integrin recruitment to rigidities associated with healthy or malignant tissue, respectively. In vitro experiments and theoretical modelling further demonstrate that this behaviour is explained by the different binding and unbinding rates of both integrin types to fibronectin. Moreover, rigidity sensing through differences in integrin bond dynamics applies both when integrins bind separately and when they compete for binding to fibronectin.


Subject(s)
Antigens, Neoplasm/metabolism , Fibronectins/metabolism , Integrins/metabolism , Mechanotransduction, Cellular/physiology , Models, Biological , Receptors, Vitronectin/metabolism , Antigens, Neoplasm/genetics , Cells, Cultured , Fibronectins/genetics , Humans , Integrins/genetics , Receptors, Vitronectin/genetics
17.
Am J Pathol ; 184(3): 842-58, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412090

ABSTRACT

Overexpression of osteopontin (OPN) is strongly associated with the invasiveness/metastasis of many cancers, including melanomas. However, the molecular mechanisms of OPN in these processes remain poorly understood. We found that forced expression of OPN in early vertical-growth-phase melanoma cells dramatically increased their migration/invasion and growth/survival in a three-dimensional collagen I gel. Neutralizing antibodies to OPN, integrin ß1, and integrin αvß3, but not to CD44, negated the effects of OPN. Conversely, knocking down OPN in metastatic melanoma cells abrogated the invasive growth. OPN overexpression activated and OPN knockdown inactivated αvß3 and αvß5 integrins, negligibly affecting their expression. We further found OPN expression to inversely correlate with tetraspanin CD9 expression. Early-stage melanoma cells displayed low OPN and high CD9 expression, and conversely, metastatic cells displayed high OPN and low CD9 expression. Overexpression of OPN in vertical-growth-phase melanoma cells induced down-regulation of CD9, and knockdown of OPN in metastatic melanoma cells up-regulated CD9. Reversion of these CD9 changes abolished the effects of OPN. Furthermore, knockdown of CD9 in early-stage melanoma cells stimulated their invasive capacity in three-dimensional collagen. Similarly, microarray analyses of benign nevi and primary melanomas from different stages revealed an inverse correlation between OPN and CD9. These data suggest that OPN promotes melanoma cell invasion by activating integrin αvß3 and down-regulating CD9, a putative metastasis suppressor.


Subject(s)
Integrin alphaVbeta3/metabolism , Melanoma/pathology , Osteopontin/metabolism , Receptors, Vitronectin/metabolism , Tetraspanin 29/metabolism , Cell Line, Tumor , Cell Movement , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Integrin alphaVbeta3/genetics , Melanoma/genetics , Melanoma/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Osteopontin/genetics , Receptors, Vitronectin/genetics , Tetraspanin 29/genetics , Up-Regulation
18.
Toxicol Appl Pharmacol ; 289(1): 20-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26344001

ABSTRACT

Connective tissue growth factor (CTGF; also known as CCN2) is an inflammatory mediator that is abundantly expressed in osteoarthritis (OA). Interleukin-1ß (IL-1ß) plays a pivotal role in OA pathogenesis. Berberine exhibits an anti-inflammatory effect, but the mechanisms by which it modulates CCN2-induced IL-1ß expression in OA synovial fibroblasts (OASFs) remain unknown. We showed that CCN2-induced IL-1ß expression is mediated by the activation of αvß3/αvß5 integrin-dependent reactive oxygen species (ROS) generation, and subsequent activation of apoptosis signal-regulating kinase 1 (ASK1), p38/JNK, and nuclear factor-κB (NF-κB) signaling pathways. This IL-1ß expression in OASFs is attenuated by N-acetylcysteine (NAC), inhibitors of ASK1, p38, or JNK, or treatment with berberine. Furthermore, berberine also reverses cartilage damage in an experimental model of collagenase-induced OA (CIOA). We observed that CCN2 increased IL-1ß expression via αvß3/αvß5 integrins, ROS, and ASK1, p38/JNK, and NF-κB signaling pathways. Berberine was found to inhibit these signaling components in OASFs in vitro and prevent cartilage degradation in vivo. We suggest a novel therapeutic strategy of using berberine for managing OA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Berberine/pharmacology , Connective Tissue Growth Factor/pharmacology , Interleukin-1beta/metabolism , Osteoarthritis/drug therapy , Acetylcysteine/pharmacology , Animals , Cartilage/drug effects , Cartilage/metabolism , Cells, Cultured , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Interleukin-1beta/genetics , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Stem Cells ; 32(2): 327-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24123709

ABSTRACT

An imbalance between normal adipogenesis and osteogenesis by mesenchymal stem cells (MSCs) has been shown to be related to various human metabolic diseases, such as obesity and osteoporosis; however, the underlying mechanisms remain elusive. We found that the interaction between osteopontin (OPN), an arginine-glycine-aspartate-containing glycoprotein, and integrin αv/ß1 plays a critical role in the lineage determination of MSCs. Although OPN is a well-established marker during osteogenesis, its role in MSC differentiation is still unknown. Our study reveals that blockade of OPN function promoted robust adipogenic differentiation, while inhibiting osteogenic differentiation. Re-expression of OPN restored a normal balance between adipogenesis and osteogenesis in OPN(-/-) MSCs. Retarded bone formation by OPN(-/-) MSCs was also verified by in vivo implantation with hydroxyapatite-tricalcium phosphate, a bone-forming matrix. The role of extracellular OPN in MSC differentiation was further demonstrated by supplementation and neutralization of OPN. Blocking well-known OPN receptors integrin αv/ß1 but not CD44 also affected MSC differentiation. Further studies revealed that OPN inhibits the C/EBPs signaling pathway through integrin αv/ß1. Consistent with these in vitro results, OPN(-/-) mice had a higher fat to total body weight ratio than did wild-type mice. Therefore, our study demonstrates a novel role for OPN-integrin αv/ß1 in regulating MSC differentiation.


Subject(s)
Adipogenesis/genetics , Osteogenesis/genetics , Osteopontin/metabolism , Receptors, Vitronectin/metabolism , Adipocytes/cytology , Animals , Cell Differentiation/drug effects , Cell Lineage , Humans , Mesenchymal Stem Cells , Mice , Osteoblasts/metabolism , Osteopontin/genetics , Protein Interaction Maps/genetics , Receptors, Vitronectin/genetics
20.
J Biol Chem ; 288(17): 12366-75, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23504323

ABSTRACT

Heparanase functions as a heparan sulfate-degrading enzyme and as a ligand for an unidentified signaling receptor(s). Here, several reactions involved in the activation of the PI3K-AKT pathway by latent heparanase were characterized. Protein suppression using specific siRNAs revealed that heparanase-induced phosphorylation of AKT at Ser-473 was RICTOR-mTOR-dependent, whereas ILK and PAK1/2 were dispensable. p110α was the PI3K catalytic isoform preferred by heparanase for AKT activation and cell proliferation because the p110α inhibitor YM024 blocked these processes. Heparanase-induced AKT phosphorylation was low in mouse embryonic fibroblast cells expressing a RAS interaction-defective p110α compared with wild type cells, indicating that RAS has an important role in the PI3K-AKT activation. The response to heparanase was also inefficient in suspension cultures of several cell lines, suggesting a requirement of integrins in this pathway. Adhesion via either αVß3 or α5ß1 promoted heparanase-induced AKT phosphorylation, and a stronger effect was seen when both integrins were engaged. Simultaneous inhibition of FAK and PYK2 using a chemical inhibitor, or suppression of their expression, inhibited heparanase-induced AKT activation and cell proliferation. Stimulation of cells with heparanase enhanced their resistance against oxidative stress- or growth factor starvation-induced apoptosis. These results demonstrate that there is an intimate cross-talk between the heparanase receptor(s) and integrins during induction of the prosurvival PI3K-AKT pathway by heparanase.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Glucuronidase/metabolism , Integrin alphaVbeta3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Vitronectin/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Cricetinae , Cricetulus , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Glucuronidase/genetics , Humans , Integrin alphaVbeta3/genetics , Mice , Morpholines/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/genetics , Receptors, Vitronectin/genetics , Signal Transduction/drug effects , Thiophenes/pharmacology , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL