crossmark THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 290, NO. 42, pp. 25534 –25547, October 16, 2015 © 2015 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

Integrin ␣3␤1 Binding to Fibronectin Is Dependent on the Ninth Type III Repeat* Received for publication, April 3, 2015, and in revised form, August 13, 2015 Published, JBC Papers in Press, August 28, 2015, DOI 10.1074/jbc.M115.656702

Ashley C. Brown‡, Marilyn M. Dysart§, Kimberly C. Clarke¶, Sarah E. Stabenfeldt储, and Thomas H. Barker§**1 From the ‡Department of Biomedical Engineering, North Carolina State University and the University of North Carolina, Raleigh, North Carolina 27606, the §Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta Georgia 30332, the ¶School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, the 储School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, and the **Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332 Background: The fibronectin (Fn) ninth type III repeat can modulate integrin binding and resulting cell spreading. Results: Mutations within the Fn integrin binding domains affect integrin ␣3␤1 binding. Conclusion: Integrin ␣3␤1-fibronectin binding depends on the presence and spacing of the RGD and synergy sites within Fn. Significance: ␣3␤1-fibronectin binding may modulate epithelial cell wound healing responses. Fibronectin (Fn) is a promiscuous ligand for numerous cell adhesion receptors or integrins. The vast majority of Fn-integrin interactions are mediated through the Fn Arg-Gly-Asp (RGD) motif located within the tenth type III repeat. In the case of integrins ␣IIb␤3 and ␣5␤1, the integrin binds RGD and the synergy site (PHSRN) located within the adjacent ninth type III repeat. Prior work has shown that these synergy-dependent integrins are exquisitely sensitive to perturbations in the Fn integrin binding domain conformation. Our own prior studies of epithelial cell responses to recombinant fragments of the Fn integrin binding domain led us to hypothesize that integrin ␣3␤1 binding may also be modulated by the synergy site. To explore this hypothesis, we created a variety of recombinant variants of the Fn integrin binding domain: (i) a previously reported (Leu 3 Pro) stabilizing mutant (FnIII9ⴕ10), (ii) an Arg to Ala synergy site mutation (FnIII9R3A10), (iii) a two-Gly (FnIII92G10) insertion, and (iv) a four-Gly (FNIII94G10) insertion in the interdomain linker region and used surface plasmon resonance to determine binding kinetics of integrin ␣3␤1 to the Fn fragments. Integrin ␣3␤1 had the highest affinity for FnIII9ⴕ10 and FnIII92G10. Mutation within the synergy site decreased integrin ␣3␤1 binding 17-fold, and the four-Gly insertion decreased binding 39-fold compared with FnIII9ⴕ10. Cell attachment studies demonstrate that ␣3␤1-mediated epithelial cell binding is greater on FnIII9ⴕ10 compared with the other fragments. These studies suggest that the presence and spacing of the RGD and synergy sites modulate integrin ␣3␤1 binding to Fn.

Cells interact with their surrounding extracellular matrix (ECM)2 via transmembrane cell surface receptors, known as integrins. Integrins are heterodimeric proteins consisting of

* This work was supported, in whole or in part, by National Institutes of Health Grants R01EB011566 and R01HL127283. The authors declare that they have no conflicts of interest with the contents of this article. To whom correspondence should be addressed: Wallace H. Coulter Dept. of Biomedical Engineering at Georgia Tech and Emory University, 950 Atlantic Dr., Ste. 4015, Atlanta, GA 30332. Tel.: 404-385-5039; Fax: 404-894-4243; E-mail: [email protected]. 2 The abbreviations used are: ECM, extracellular matrix; Fn, fibronectin; SPR, surface plasmon resonance; RSS, residual sum of squares; Ln, laminin. 1

25534 JOURNAL OF BIOLOGICAL CHEMISTRY

one ␣ and one ␤ subunit, which are known to form at least 24 unique heterodimers (1). Integrin interactions with their ECM ligands facilitate a host of cellular responses, including cell spreading, migration, proliferation, and differentiation, and can contribute to more orchestrated cellular events, such as angiogenesis and epithelial to mesenchymal transitions, among others (2– 4). Integrin binding to ECM ligands occurs through specific binding sequences, the most notable of these sequences being Arg-Gly-Asp (RGD), which is found on a large number of ECM proteins, including fibronectin, vitronectin, osteoponin, laminin, thrombospondin, and several others (5). Furthermore, integrin heterodimers can interact with multiple ECM ligands and bind to multiple binding sequences (6, 7). Fibronectin (Fn) is a widely expressed ECM protein and is known to bind at least 16 integrins. Biochemically, Fn exists as a soluble dimeric glycoprotein composed of two nearly identical 230 –270-kDa monomers linked covalently near their C termini by a pair of disulfide bonds (8, 9). Each monomeric subunit consists of three types of repeating modules: types I, II, and III. These modules comprise functional domains that mediate interactions with other ECM components, cell surface receptors, and Fn itself (9). Whereas type I and II repeats are structurally stabilized with two intrachain disulfide bonds in each repeat, type III repeats have no disulfide bonds and therefore are highly sensitive to external stimuli, including application of force, resulting in alterations of conformation of the molecule (8, 10 –12). Interestingly, a large number of Fn-integrin interactions occur through the RGD site located on the tenth type III repeat. The recognition of this simple tripeptide sequence can be quite complex and greatly depends on flanking residues, its three-dimensional presentation, and individual features of the integrin-binding pockets. This dependence is most well characterized in ␣5␤1 integrin binding to Fn, where RGD in concert with a second recognition sequence (PHSRN), the “synergy” site, in the adjacent ninth type III repeat is believed to promote the specific interaction of ␣5␤1 integrin binding to Fn through interactions with the ␣5 subunit (13–15). The synergy site is VOLUME 290 • NUMBER 42 • OCTOBER 16, 2015

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9 TABLE 1 FnIII9ⴕ10 variants produced and function Fragment FnIII9⬘10 FnIII9

R-A

10

2G

FnIII9 10 4G

FnIII9 10

Sequence 1373–1423 of FnIII9ⴕ10 DRVPHSRNSITLTNLTPGTEYVVSIVALN GREESPPLIGQQSTVSDVRPD DRVPHSANSITLTNLTPGTEYVVSIVALN GREESPPLIGQQSTVSDVRPD DRVPHSRNSITLTNLTPGTEYVVSIVALN GREESPPLIGQQSTVSGGDVRPD DRVPHSRNSITLTNLTPGTEYVVSIVALN GREESPPLIGQQSTVSGGGGDVRPD

located ⬃32 Å from the RGD loop on the tenth type III repeat. The type III repeats show great elasticity in the loops between their F- and G-␤ strands, known as the FG loop, which allows the ninth and tenth type III repeats to present multiple conformations. Under small applied forces (on the order of 10 pN), the Fn tenth type III repeat is susceptible to partial unfolding. Computational models of force application to the Fn tenth type III repeat suggest that the RGD loop within the tenth type III repeat translocates away from the ninth type III repeat, resulting in an increase in the distance between the RGD and synergy sites from ⬃32 to ⬃55 Å (16). This capacity to present multiple spatial orientations of the ninth and tenth type III repeats has implications on cell binding, because the relative positioning of these two domains has been shown to influence binding of integrins such as ␣IIb␤3 and ␣5␤1 (17–21). Further evidence suggesting that alternate conformational states induce integrin “switching” comes from studies in which conformational stability was conferred to the ninth or tenth type III repeats, resulting in modulation of integrin affinity. In these studies, the ninth type III repeat was stabilized via a Leu-Pro mutation at amino acid 1408 (22, 23), or the tenth type III repeat was stabilized through facilitation of greater hydrogen bonding within the repeat (24). In both cases, stabilization of the relative positions of the two repeats resulted in increased affinity for integrin ␣5␤1 over integrin ␣v␤3. In addition, studies in which the linker region between the ninth and tenth type III repeats was increased in length showed reduction in ␣5␤1 binding (17). Previous studies in our lab using recombinant Fn fragments displaying the RGD and synergy sites with a stabilizing (L1408P) point mutation (FnIII9⬘10) or RGD alone (FnIII10) suggest that integrin ␣3␤1 binding to Fn may also be promoted by the ninth type III repeat (26). Although the classical ligand for integrin ␣3␤1 is laminin (Ln) (27, 28), it has been reported to bind collagen and Fn (9, 29, 30) and facilitate cell-cell interactions through both homophilic binding and binding to E-cadherin (31–33). Furthermore, integrin ␣3␤1 binding to Fn has been shown to occur in an RGD-dependent fashion; however, ␣3␤1 binding to Ln and collagen does not occur through RGD sites (29). Integrin ␣3␤1 is highly expressed by many epithelial cells, including alveolar epithelial cells, renal epithelial cells, and keratinocytes, and has been shown to be critical for regulation of epithelial phenotype (34, 35). Integrin ␣3␤1 plays an important role in maintaining epithelial integrity and facilitating wound repair responses, and if not bound to its ECM ligands, it can contribute to pathologies through induction of epithelial to mesenchymal transitions (34, 36). During wound OCTOBER 16, 2015 • VOLUME 290 • NUMBER 42

Role Leu

1408

to Pro; stabilizes relative positions of FnIII9 and FnIII10

Arg to Ala point mutation in PHSRN site Increased distance between FnIII9 and FnIII10 Increased distance between FnIII9 and FnIII10

healing responses, the ECM rapidly undergoes changes in composition from mostly Ln and elastin to higher concentrations of provisional matrix components, such as Fn. Understanding the ability of integrin ␣3␤1 to bind to both its classical ligand Ln as well as Fn is important to understand cell interactions with these dynamic matrices observed in wound healing and may elucidate mechanisms involved in normal versus pathological wound healing responses. To explore the hypothesis that ␣3␤1 binding to Fn is enhanced by the presence of the ninth type III repeat and its spacing relative to the tenth type III repeat, we created (i) a dominant negative Arg-Ala mutation in the synergy site (FnIII9R-A10), (ii) a two-Gly insertion in the linker region (FnIII92G10), or (iii) a four-Gly insertion in the linker region (FnIII94G10). Surface plasmon resonance (SPR) was then utilized to determine the binding kinetics of integrin ␣3␤1 to the Fn fragments, and cell attachment and spreading assays were then utilized to validate the binding data.

Experimental Procedures Construction of Mutant pGEX4T1-FnIII9⬘10 Clones— Cloning of the Leu1408 to Pro mutation (FnIII9⬘10) was made using a parent pGEX4T-1-FN III9 –10 encoding plasmid as previously described (22, 23, 26). FnIII9⬘10 variants displaying (i) a dominant negative Arg-Ala mutation in the synergy site (FnIII9R-A10), (ii) a 2⫻ Gly insertion in the linker region (FnIII92G10), or (iii) a 4⫻ Gly insertion in the linker region (FnIII94G10) were then created using the QuikChange威 II-E site-directed mutagenesis kit (Stratagene, La Jolla, CA). Sequences are presented in Table 1. All plasmids were introduced into and maintained in the electrocompetent XL-1 Blue Escherichia coli strain and cultured on agarose with LB and ampicillin (0.1 mg/ml) at 37 °C. Plasmids were extracted from cultures using the QIAquick spin miniprep kit (Qiagen) and verified via sequencing (Johns Hopkins Synthesis & Sequencing Facility, Baltimore, MD). Expression and Purification of Recombinant FnIII9⬘10 Proteins—Recombinant Fn fragments were produced as previously described (23, 26). Briefly, the expression vectors described above were transformed into BL21 E. coli, and cells were grown to the exponential growth phase and treated with isopropyl ␤-D-thiogalactopyranoside for 3 h. Cells were then lysed by the addition of 10 mg/ml lysozyme and sonication, followed by incubation with 1% Triton X-100 and 10 units/ml of DNase I. Fn fragments were purified by GST affinity chromatography (AKTA Purifier; GE Healthcare). GST tags were removed using bovine thrombin (Sigma-Aldrich). A second round of purification was performed using GST and serine proJOURNAL OF BIOLOGICAL CHEMISTRY

25535

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9 tease affinity chromatography to remove cleaved GST tags and thrombin. Proteins were verified as ⬎98% pure by SDS-PAGE (see Fig. 1). Circular Dichroism—CD spectra were collected on a JASCOJ810 spectropolarimeter. Proteins were desalted and concentrated in DI water using PALL centrifugation tubes (3-kDa molecular mass cutoff). Aliquots (0.25 mg/ml) were placed in a quartz cuvette (1-mm path length) and scanned at 50 nm/min. The temperature was maintained at 20 °C. Surface Plasmon Resonance Studies—A Biacore 2000 (Biacore Lifesciences, GE Healthcare) was used to investigate kinetic binding constants (ka and kd) of Fn fragments variants for integrin ␣3␤1 and control integrins ␣5␤1 and ␣v␤3. Briefly, integrins (R & D Systems, Minneapolis, MN) were covalently immobilized to gold-coated SPR sensor chips via self-assembled monolayer surface chemistry to generate a nonfouling surface with a controlled density of reactive carboxylic acid groups. Mixed self-assembled monolayers were generated on goldcoated chips as previously described (37, 38) by incubating with a 10:1 mixture of 1 mM of tri(ethylene glycol)-terminated alkanethiols (HS-(CH2)11–(OCH2CH2)3-OH (ProChimia, Gdansk, Poland) and carboxylic acid-terminated alkanethiols (HS-(CH2)11–(OCH2CH2)6-OCH2COOH) overnight. The senor chip was then loaded into the Biacore 2000, and the carboxylic acid-terminated alkanethiol surface was activated by flowing 200 mM 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (Sigma-Aldrich) and 50 mM N-hydroxysuccinimide (Sigma-Aldrich; 5 ␮l/min for 10 min). Immediately after activation, integrins (100 ␮g/ml) were flowed through the device and allowed to react with the functionalized surfaces at a flow rate of 5 ␮l/min for 10 min to achieve ⬃1500 resonance units (1 resonance unit, ⬃1 pg/mm2). An additional channel was immobilized with BSA to serve as a reference channel; BSA (100 ␮g/ml) was likewise immobilized to achieve ⬃1500 resonance units. Immobilization of integrin and BSA were performed in 0.1 M sodium acetate, pH 4.5. Unreacted N-hydroxysuccinimide groups were quenched in all flow cells with 1 M ethanolamine, pH 8.5 (10 ␮l/min for 10 min). Upon stabilization of the baseline signal, kinetic binding experiments were run with Fn fragments, full-length Fn, or Ln as the flow analytes. Various concentrations for each Fn fragment (10 ␮M to 1 nM) were flowed at 30 ␮l/min for 5 min immediately followed by a 10-min dissociation phase. Between each injection, the surface was regenerated with two 30-s pulses (10 ␮l/min) of 20 mM EDTA and 1 M NaCl (pH 6.0). ␣3␤1 binding experiments were performed in 10 mM HEPES, 150 mM NaCl, 0.0001% Triton X-100, and 2 mM each MgCl2 and MnCl2, pH 7.4; ␣5␤1 and ␣v␤3 experiments were performed in this same buffer without Triton X-100. ␣3␤1 integrin interactions were also analyzed in the presence of free 100 ␮g/ml RGD peptides. To determine the role of metal ions in Fn fragment-␣3␤1integrin interactions, experiments were performed in 10 mM HEPES, 150 mM NaCl, 0.0001% Triton X-100, and 2 mM MgCl2/2 mM CaCl2. The sensor chips in different experiments were freshly immobilized with integrin (␣3␤1, ␣5␤1, or ␣v␤3). A single experiment consisted of flowing various concentrations of FnIII9⬘10 variants, Fn, or Ln over the immobilized integrin and control channel. Between each injection, the surface was regenerated. At least three inde-

25536 JOURNAL OF BIOLOGICAL CHEMISTRY

pendent experiments were performed per integrin. For replicate experiments, FnIII9⬘10 variant, Fn, and Ln injection order was varied to rule out binding trends associated with injection sequence, such as potential decreased activity of the immobilized ligand following regeneration. To characterize binding to integrin ␣3␤1 in the presence of CaCl2 and RGD, independent experiments were performed with freshly immobilized integrin. SPR Analysis and Evaluation—SPR sensorgrams were analyzed with Scrubber 2 and ClampXP software (Center for Biomolecular Interactions Analysis, University of Utah) (39 – 41). Reference cell responses were subtracted from corresponding active response curves. The resulting curves were then analyzed and fit to the kinetic models. Kinetic modeling and simulations were performed with ClampXP software with the heterogeneous surface model; globally fitted parameters were determined for each kinetic data set per Fn fragment. Equilibrium binding constants (Kd1 and Kd2) were calculated from fit kinetic constants by dividing kd/ka for each experiment; mean Kd1/2 values are presented in data tables. Goodness of fit for each model was evaluated by analyzing the residual plots and residual sum of squares. Cell Attachment Assays—Wells of a 96-well plate were coated with FnIII9⬘10 (2 ␮M), FnIII9R-A10 (2 ␮M), FnIII92G10 (2 ␮M), FnIII94G10 (2 ␮M), Fn (0.1 ␮M), or Ln (0.1 ␮M) at 4 °C and then blocked with heat-denatured BSA. The concentration of Fn was chosen based on previous studies showing similar binding of an antibody specific to the 7–10 type III repeats of Fn (clone HFN7.1a1) (23) to Fn coated at 0.1 ␮M and the Fn fragments at 2 ␮M concentrations. The concentration of Ln was chosen based on ELISAs showing saturation of the surface using 0.1 ␮M. RLE-6TN cells were incubated in microcentrifuge tubes at a concentration of 3 ⫻ 106 cells/ml in DMEM with or without the addition of anti-␣3 (clone H-43), ␣5 (clone A-11), or ␣v (clone H-75) blocking antibodies or combinations of the three for 30 min at 37 °C. All antibodies were purchased from Santa Cruz Biotechnology (Dallas, TX). To determine 100% attachment, RLE-6TN cells were plated on poly-L-lysine-coated tissue culture plastic wells. Cells with or without the addition of antibodies were then plated on each of the Fn fragment-, Fn-, or Ln-coated wells for 30 min, and then all control and treated wells were washed and fixed with 5% gluteraldehyde and stained with 0.1% crystal violet stain. The dye was solubilized in 10% acetic acid, and absorbance was measured on a Biotek Synergy H4 multi-mode plate reader. The results are pooled from three independent triplicate experiments and presented as a percentage of attachment with respect to the poly-L-lysine signal. Epithelial Cell Spreading on FnIII9⬘10 Variants—The influence of FnIII9⬘10 variants on epithelial cell spreading was analyzed by culturing RLE-6TN cells on FnIII9⬘10(2 ␮M), FnIII9R-A10- (2 ␮M), FnIII92G10- (2 ␮M), FnIII94G10(2 ␮M), Fn- (0.1 ␮M), or Ln-coated (0.1 ␮M), hd-BSA blocked coverslips in serum-free DMEM/F12 medium. To determine the effect of various integrins on cell spreading, cells were incubated with ␣3, ␣5, or ␣v blocking antibodies at 37 °C for 30 min prior to plating. Following a 3-h incubation period, VOLUME 290 • NUMBER 42 • OCTOBER 16, 2015

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

FIGURE 1. FnIII9ⴕ10 variant analysis and integrin ␣3␤1 immobilization. A and B, SDS-PAGE gel (A) and CD spectra (B) of purified FnIII9⬘10 variants are presented. C, to analyze integrin binding to FnIII9⬘10 variants, recombinant soluble integrin was immobilized onto a Biacore sensor chip to achieve ⬃1500 resonance units. D, schematic of immobilized integrins on surface. Amine coupling can result in random orientation of immobilized integrins.

the cells were washed with PBS, fixed with 4% formaldehyde, permeabilized with 0.2% Triton X-100, and then blocked with 10% goat serum. Polymerized actin was stained with Alexa Flour 546 phalloidin (Invitrogen), and nuclei were stained with Hoescht stain (Invitrogen). Coverslips were mounted, and images were examined using confocal microscopy (40⫻ oil immersion objective; Zeiss 700 – 405). RepreOCTOBER 16, 2015 • VOLUME 290 • NUMBER 42

sentative images are presented. The area and perimeter of individual cells were determined for each condition using ImageJ (NIH Freeware) image processing software, and circularity was determined using the equation circularity ⫽ 4␲(area/perimeter2). At least 50 cells were analyzed per condition. The cumulative frequency distribution of area and circularity of cells was determined for each data set. JOURNAL OF BIOLOGICAL CHEMISTRY

25537

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9 TABLE 2 Kd values: ␣3␤1-heterogeneous surface model Kinetic association (ka) and dissociation (kd) rates obtained from the fits are presented. ka1 and kd1 are associated with the high affinity binding state, and ka2 and kd2 are associated with the low affinity binding state. Equilibrium dissociation constants, which take kd and ka values into account, were calculated for each analyte for the high (Kd1) and low (Kd2) affinity binding state. RSS are also presented. Analyte

ka1 ⫻ 104

kd1 ⫻ 10ⴚ4

Kd1 ⫻ 10ⴚ9

FnIII9⬘10 Fn9R-A10 FnIII92G10 FnIII94G10 Fn Ln

333 ⫾ 294 70.2 ⫾ 67.4 1529 ⫾ 1520 15.8 ⫾ 13.2 15.6 ⫾ 14.1 243 ⫾ 239

136 ⫾ 89.9 471 ⫾ 276 492 ⫾ 488 247 ⫾ 241 2.43 ⫾ 1.6 228 ⫾ 226

4.1 67.2 3.2 156 1.6 9.4

Statistical Analysis—All statistical analyses were performed with Prism software program (GraphPad, San Diego CA). The data were analyzed using a one-way analysis of variance using the Tukey test at a 95% confidence interval.

Results Analysis of FnIII9⬘10 Variants—FnIII9⬘10 variants were analyzed by SDS-PAGE and CD (Fig. 1). Variants were verified as ⬎98% pure by SDS-PAGE. CD spectra of all FnIII9⬘10 variants are in good agreement with that of full-length fibronectin (42) (i.e. minimum at 215 nm and maxima at 226 and 202 nm), except for fragment FNIII94G10, which has a minimum of ⬃216 nm and a maximum of ⬃194 nm. These bands are suggestive of ␤-sheets. Similar changes in CD spectra for FNIII9 –10 have been observed upon partial unfolding (43, 44). The additional glycine residues within the loop region may have resulted in a more open conformation of the FNIII94G10 fragment, analogous to partial unfolding of the domain. However, the secondary structures of the fragments clearly remain intact, because spectra for unfolded, random coils are not present. Fitted ␣3␤1 Binding Affinity Parameters: Heterogeneous Surface Model—SPR with soluble recombinant integrin immobilized to the surface of the sensor chip was utilized to determine the role of FnIII9⬘10 in integrin ␣3␤1 interactions with Fn (Fig. 1). A representative sensorgram obtained from integrin ␣3␤1 immobilization is shown in Fig. 1C. The large spike in RU seen following injection of ethanolamine is due to differences in refractive indices between the immobilization buffer and ethanolamine. Experimental data were collected and then fit to a two-site heterogeneous surface model to account for variable orientation and conformational state of immobilized integrin. Integrins inherently can present both high and low affinity states, which can result in a heterogeneous surface displaying complex binding kinetics. Furthermore, the amine coupling procedure utilized for immobilization of integrin onto the surface can result in a random receptor orientation (Fig. 1D) and a heterogeneous surface, also presenting both high and low affinity states of receptor. To account for the possibility of both high and low affinity states of the integrin, a heterogeneous surface model was utilized to fit the response curves. The fitted parameters ka1, kd1, ka2, kd2, calculated Kd1 and Kd2, and residual sum of squares of the fit of each FnIII9⬘10 variant, Fn, and Ln using a heterogeneous surface model are displayed in Table 2. Experimental response and fit simulation curves are presented for FnIII9⬘10 variants binding to integrin ␣3␤1 in Fig. 2A. Residual

25538 JOURNAL OF BIOLOGICAL CHEMISTRY

ka2 ⫻ 104

kd2 ⫻ 10ⴚ4

0.25 ⫾ 0.1 2.59 ⫾ 2.4 0.43 ⫾ 0.4 0.01 ⫾ 0.01 1.2 ⫾ 1.1 13.6 ⫾ 11.7

214 ⫾ 201 883 ⫾ 650 49.4 ⫾ 38.3 62.3 ⫾ 35.4 2183 ⫾ 1415 137 ⫾ 135

Kd2 ⫻ 10ⴚ9 8427 3410 1143 45,873 2184 101

RSS 1.3 ⫾ 0.2 1.45 ⫾ 0.5 1.4 ⫾ 0.6 1.3 ⫾ 0.7 0.3 ⫾ 0.1 1.3 ⫾ 1.2

plots for FnIII9⬘10 variants fit to the heterogeneous surface model are shown in Fig. 2B. As expected, examination of the fitted parameters obtained from a heterogeneous surface model demonstrates a high affinity binding event and a secondary low affinity binding event. High affinity states of integrin result in high affinity binding events, described collectively by ka1, kd1, and the resulting calculated Kd1. Likewise, lower affinity states of the integrin result in a series of lower affinity binding events, described collectively by ka2, kd2, and the resulting calculated Kd2. It was found that FnIII9⬘10 had the lowest kd1 of all FnIII9⬘10 variants analyzed, with a value of 136 ⫻ 10⫺4 s⫺1. Increasing the linker region between the ninth and tenth type III repeats with either two Gly residues or four Gly residues, resulted in an increase in the kd to 492 ⫻ 10⫺4 and 247 ⫻ 10⫺4 s⫺1, respectively, suggesting that increasing the space between the PHSRN and RGD sites disrupts the interactions between FnIII9 and FnIII10 domains and integrin ␣3␤1. Furthermore, the disrupting Arg to Ala point mutation within the PHSRN site resulted in an increased kd of 471 ⫻ 10⫺4 s⫺1. Equilibrium dissociation constants, which takes kd1 and ka1 values into account, were calculated for each analyte for the high affinity binding state (Kd1). FnIII9⬘10 and FnIII92G10 were found to have similar Kd1 values: 4.1 and 3.2 nM, respectively. Direct disruption in the PHSRN site resulted in a 17-fold increase in Kd to 67.2 nM (compared with FnIII9⬘10), whereas increasing the spacing between the ninth and tenth type III repeats by four Gly residues resulted in a 39-fold increase to 156 nM. Ln and Fn analytes were found to have a high affinity for the integrin as well, with a Kd of 9.4 and 1.6 nM, respectively. Experimental response and fit simulation curves for Fn and Ln binding to integrin ␣3␤1 are shown in Figs. 3 and 4, respectively. Focusing on the high affinity binding parameters obtained from the heterogeneous surface model demonstrates that stabilization of the ninth and tenth type III domains of Fn results in affinities for integrin ␣3␤1 similar to those observed for the classical ␣3␤1 ligand Ln. These results indicate that Fn does bind integrin ␣3␤1 with a high affinity, and this binding is dependent on the spacing of the PHSRN and RGD sites. Integrin binding is typically dependent on the presence of divalent cations (29, 45), and so all SPR experimental samples were performed in the presence of 2 mM of MgCl2 and MnCl2. To confirm that integrin binding to FnIII9⬘10 variants is dependent on the presence of divalent cations, binding events were characterized in the presence of 2 mM MgCl2 and CaCl2, because manganese has been known to actiVOLUME 290 • NUMBER 42 • OCTOBER 16, 2015

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

FIGURE 2. FnIII9ⴕ10 variant binding to immobilized integrin ␣3␤1. A, response curves obtained from FnIII9⬘10 variants binding to immobilized integrin ␣3␤1 were fit to a heterogeneous surface model. Experimental response curves and the resulting fits are shown for FnIII9⬘10, FnIII9R-A10, FnIII92G10, and FnIII94G10. Solid lines indicate experimental SPR response curves, and dashed curves indicate fitted models. B, representative residual plots are shown for FnIII9⬘10, FnIII9R-A10, FnIII92G10, and FnIII94G10.

OCTOBER 16, 2015 • VOLUME 290 • NUMBER 42

JOURNAL OF BIOLOGICAL CHEMISTRY

25539

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

FIGURE 3. Fibronectin binding to integrin ␣3␤1. Response curves obtained from Fn binding to immobilized integrin ␣3␤1 were fit to a heterogeneous surface model. A, experimental response curves and resulting fits are shown for a range of Fn concentrations. Solid lines indicate experimental SPR response curves, and dashed curves indicate fitted models. B, residuals.

vate integrin by occupying the metal ion binding sites of integrin. In the presence of CaCl2 instead of MnCl2, minimal binding to integrin ␣3␤1 was observed (Fig. 5A), indicating as expected, that ␣3␤1 binding is dependent on the presence of divalent cations. To determine whether the RGD site of FN is critical for the observed integrin interactions, binding of FnIII9⬘10 variants to integrin ␣3␤1 was analyzed in standard running buffer (2 mM of MgCl2 and MnCl2) in the presence of 100 ␮g/ml of RGD peptide (Fig. 5B). Binding was inhibited in the presence of RGD, indicating that RGD is critical for integrin ␣3␤1 binding to FnIII9⬘10 variants. Control Integrins: Fitted ␣5␤1 and ␣v␤3 Binding Affinity Parameters—The role of the PHSRN site, as well as the relative spacing with respect to the RGD site, is critical for ␣5␤1 binding to Fn, whereas binding to ␣v␤3 only requires the RGD site (46). To confirm these previous reports and provide validation of our methods, binding of FnIII9⬘10, FnIII9R-A10, FnIII92G10, FnIII94G10, Fn, and Ln to immobilized integrin ␣5␤1 and ␣v␤3 was investigated. As with ␣3␤1, curves were fit to a heterogeneous surface model. Representative response curves and heterogeneous surface model fits for binding of one concentration (2 ␮M) of FnIII9⬘10 variants to integrin ␣5␤1 and ␣v␤3 are shown in Fig. 6A, and residual plots are shown in Fig. 6B. Calculated Kd values for both fits are displayed in Tables 3 and 4. As expected, ␣5␤1 binding was found to depend on the presence of the PHSRN site. Analysis of binding parameters using the het-

25540 JOURNAL OF BIOLOGICAL CHEMISTRY

FIGURE 4. Laminin binding to integrin ␣3␤1. Response curves obtained from Ln binding to immobilized integrin ␣3␤1 were fit to a heterogeneous surface model. A, experimental response curves and resulting fits are shown for a range of Ln concentrations. Solid lines indicate experimental SPR response curves, and dashed curves indicate fitted models. B, residuals.

FIGURE 5. FnIII9ⴕ10 variant binding to integrin ␣3␤1 is inhibited in the presence of MgCl2/CaCl2 buffer (A) or the presence of 100 ␮g/ml RGD peptides in MgCl2/MnCl2 buffer (B). Experimental response curves for 2 ␮M FnIII9⬘10, FnIII9R-A10, FnIII92G10, and FnIII94G10 binding to integrin ␣3␤1 are shown in MgCl2/MnCl2 running buffer (gray scale lines) or MgCl2/CaCl2 running buffer (A) or MgCl2/MnCl2 running buffer ⫹ RGD (B; colored lines).

erogeneous surface model demonstrates that the calculated Kd1 for the FnIII9⬘10 variants follow similar trends as those observed for integrin ␣3␤1; FnIII9⬘10 had the lowest Kd1 of all the variants, with a value of 41 nM. FnIII9R-A10, FnIII92G10, and FnIII94G10 were found to have Kd1 values of 735, 523, and 830 VOLUME 290 • NUMBER 42 • OCTOBER 16, 2015

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

FIGURE 6. Attachment to control integrins. Response curves obtained from FnIII9⬘10 variants binding to immobilized integrin ␣5␤1 or ␣v␤3 were fit to a heterogeneous surface model. A range of concentrations was tested for each variant. A, representative experimental response curves and resulting fits are shown for 2 ␮M FnIII9⬘10, FnIII9R-A10, FnIII92G10, and FnIII94G10. Solid lines indicate experimental SPR response curves, and dashed curves indicate fitted models. B, representative residuals are shown for FnIII9⬘10, FnIII9R-A10, FnIII92G10, and FnIII94G10.

TABLE 3 Kd values: ␣5␤1-heterogeneous surface model Kinetic association (ka) and dissociation (kd) rates obtained from the fits are presented. ka1 and kd1 are associated with the high affinity binding state, and ka2 and kd2 are associated with the low affinity binding state. Equilibrium dissociation constants, which take kd and ka values into account, were calculated for each analyte for the high (Kd1) and low (Kd2) affinity binding state. RSS are also presented. Analyte

ka1 ⴛ 104

kd1 ⴛ 10ⴚ4

FnIII9⬘10 FnIII9R-A10 FnIII92G10 FnIII94G10 Fn Ln

1.3 ⫾ 1.2 0.2 ⫾ 0.2 0.2 ⫾ 0.3 0.2 ⫾ 0.3 12.6 ⫾ 13.7 6.2 ⫾ 9.7

7.4 ⫾ 10.9 4.9 ⫾ 3.5 2.7 ⫾ 3.8 3.5 ⫾ 1.6 3.9 ⫾ 2.8 1614.2 ⫾ 2390.0

Kd1 ⴛ 10ⴚ9 41 735 523 830 86 3509

nM, respectively. Fn and Ln were found to have Kd1 values of 86 and 3509 nM, respectively. Although Ln is not typically regarded as a ligand for ␣5␤1, there have been previous reports demonstrating interactions between these molecules (47). Kd2 values were higher than the calculated Kd1 values for all analytes, ranging from 531 to 162,333 nM. FnIII9⬘10 binding to integrin ␣v␤3 was not dependent on the presence of the PHSRN site, which was expected because ␣v␤3-Fn interactions are dominated by the RGD loop. Using the heterogeneous surface model, Kd1 values for FnIII9⬘10, FnIII9R-A10, FnIII92G10, FnIII94G10, and Fn were 65, 57, 47, 30, and 82 nM, respectively, whereas minimal binding was observed to Ln. Kd2 values were considerably higher than the calculated Kd1 values for all analytes, ranging from 219 to 41,269 nM. Cell Attachment Assays—To characterize how epithelial cells attach to different FnIII9⬘10 variants, RLE-6TN alveolar epithelial cells were incubated with ␣3, ␣5, or ␣v antibodies or combinations of the three for 30 min, then plated on each of the FnIII9⬘10 variants, Fn, or Ln and allowed to attach for 30 min. OCTOBER 16, 2015 • VOLUME 290 • NUMBER 42

ka2 ⴛ 104

kd2 ⴛ 10ⴚ4

175.3 ⫾ 302.9 2.4 ⫾ 3.5 81.3 ⫾ 130.6 24.7 ⫾ 21.6 0.13 ⫾ 0.1 0.3 ⫾ 0.4

368.4 ⫾ 548.8 2872.3 ⫾ 4718.3 446.2 ⫾ 440.5 3638.7 ⫾ 3284.9 6.1 ⫾ 0.2 3284 ⫾ 4968

Kd2 ⴛ 10ⴚ9 2023 46,444 639 2707 531 162,333

RSS 1.1 ⫾ 0.5 0.8 ⫾ 0.3 1.1 ⫾ 0.3 1.6 ⫾ 0.8 4.0 ⫾ 2.8 7.6 ⫾ 8.1

This cell type was chosen because it is known to express ␣3, ␣5, and ␣v integrin subunits (26, 49); expression of ␣3 is typically approximately 10 times higher than ␣5 or ␣v expression. We have confirmed this integrin expression pattern by flow cytometry analysis (data not shown). We have previously demonstrated that attachment of RLE-6TN cells to FnIII9⬘10 is dependent on both the RGD and PHSRN sites of FN. 1 ␮g/ml of RGD peptide inhibits RLE-6TN attachment to FnIII9⬘10 by 70%, and in the presence of both 1 ␮g/ml of RGD and 1 ␮g/ml PHSRN peptides, RLE-6TN attachment to FnIII9⬘10 is completely abrogated (26). Following the 30-min incubation period, attached cells were fixed to the surface and stained, and absorbance was measured. The results shown are presented as a percentage of attachment compared with attachment on poly-Llysine. These data coincide closely with the SPR results, indicating that cell attachment to Ln and FnIII9⬘10 surfaces is highly dependent on ␣3 integrin, whereas cell attachment to FnIII92G10, FnIII94G10, and Fn was predominantly mediated by ␣v integrin (Fig. 7). Interestingly, cells cultured on the JOURNAL OF BIOLOGICAL CHEMISTRY

25541

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9 TABLE 4 Kd values: ␣v␤3-heterogeneous surface model Kinetic association (ka) and dissociation (kd) rates obtained from the fits are presented. ka1 and kd1 are associated with the high affinity binding state, and ka2 and kd2 are associated with the low affinity binding state. Equilibrium dissociation constants, which take kd and ka values into account, were calculated for each analyte for the high (Kd1) and low (Kd2) affinity binding state. RSS are also presented. Analyte

ka1 ⴛ 104

kd1 ⴛ 10ⴚ4

FnIII9⬘10 FnIII9R-A10 FnIII92G10 FnIII94G10 Fn Ln

2.5 ⫾ 0.7 3.2 ⫾ 1.9 5.7 ⫾ 3.4 9.6 ⫾ 3.8 15563.4 ⫾ 14046.6

15.7 ⫾ 6.5 12.8 ⫾ 5.4 13.1 ⫾ 1.7 10.6 ⫾ 2.6 3471.9 ⫾ 2535.3

Kd1 ⴛ 10ⴚ9 65 57 47 30 82

ka2 ⴛ 104

kd2 ⴛ 10ⴚ4

5.7 ⫾ 3.5 5.3 ⫾ 3.4 41.9 ⫾ 21.2 0.9 ⫾ 0.4 104.4 ⫾ 104.3

183.5 ⫾ 64.3 101 ⫾ 70.0 648.7 ⫾ 250.3 114.4 ⫾ 45.0 10.0 ⫾ 4.4

Kd2 ⴛ 10ⴚ9 41,269 219 432 2157 509

RSS 5.2 ⫾ 1.9 2.3 ⫾ 1.1 3.7 ⫾ 0.9 5.4 ⫾ 2.8 3.5 ⫾ 0.9

FIGURE 7. Attachment assays on Fn variants. RLE-6TN alveolar epithelial cells were incubated with ␣3, ␣5, or ␣v blocking antibodies and cultured on each of the FnIII9 –10 variants, Fn, or Ln. The results are pooled from three independent triplicate experiments and are reported as a percentage of 100% attachment. ***, p ⬍ 0.001; **, p ⬍ 0.01; *, p ⬍ 0.05.

FnIII9R-A10 surface appear to mediate attachment through both ␣3 and ␣5 integrins. Previous studies have suggested that the RGD and PHSRN sites could serve as an on/off switch for ␣5␤1 binding to Fn such that decoupling of the PHSRN and

25542 JOURNAL OF BIOLOGICAL CHEMISTRY

RGD sites may turn the switch “off” for ␣5␤1 binding and “on” for ␣v integrin binding. Although this is supported by our results with the two-Gly and four-Gly insertion variants, it appears that a mutation in the synergy site is not sufficient to drive epitheVOLUME 290 • NUMBER 42 • OCTOBER 16, 2015

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

FIGURE 8. Cell spreading on FnIII9ⴕ10 variants. A, RLE-6TN alveolar epithelial cells were analyzed for cell spreading responses on FnIII9⬘10, FnIII9R-A10, FnIII92G10, FnIII94G10, Fn, and Ln following a 3-h incubation period. Scale bar, 20 ␮m. Actin (red) was visualized through staining with Alexa Flour 546 phalloidin. B, spread area and circularity was calculated to quantify differences in cell spreading/shape. ***, p ⬍ 0.001; **, p ⬍ 0.01; *, p ⬍ 0.05. C, cumulative frequency distribution of cell area and circularity is presented to demonstrate shifts in the distribution of cell area and circularity on these substrates.

lial cells to predominantly attach through ␣v integrin over ␣3 and ␣5 integrins. Overall, these results indicate that stabilization of the RGD and synergy sites in the FnIII9⬘10 variant facilitate cell attachment predominantly through ␣3 integrins, whereas increasing the linker region in FnIII92G10 and FnIII94G10 drives cell attachment predominantly through ␣v integrins. Epithelial Cell Spreading on FnIII9⬘10 Variants—Spreading of epithelial cells on FnIII9⬘10 variants was analyzed by plating RLE-6TN cells on FnIII9⬘10 variants, Fn, or Ln for 3 h then subsequently staining for polymerized actin (Fig. 8). Cell spread area was analyzed on the various substrates. Spreading was OCTOBER 16, 2015 • VOLUME 290 • NUMBER 42

found to be greatest on Ln and Fn. On FnIII9⬘10 variants, cell spread area was greatest on FnIII9⬘10 and least spread on FnIII92G10; however, these differences in spread area on the variants were not significantly different. Furthermore, inhibition of integrin with antibodies did not significantly change cell spreading on FnIII9⬘10 variants (Fig. 9). Differences in cell shape were additionally analyzed by calculating cell circularity, with values closer to 1 indicating a more rounded cell. Cell circularity has previously been utilized to characterize epithelial to mesenchymal responses of alveolar epithelial type II cells (2, 3, 26). In these previous studies, cells that engage ␣3 integrins have been JOURNAL OF BIOLOGICAL CHEMISTRY

25543

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

FIGURE 9. Cell area on FnIII9ⴕ10 variants in the presence of integrin blocking antibodies. To determine the influence of integrin engagement on RLE-6TN cell spreading responses on FnIII9⬘10 variants, RLE-6TN cells were incubated with ␣3, ␣5, or ␣v antibodies for 30 min prior to plating on FnIII9⬘10, FnIII9R-A10, FnIII92G10, FnIII94G10, Fn, and Ln. Cumulative frequency distribution of cell area was utilized to visualize shifts in the distribution of cell area on these substrates in the presence of integrin blocking antibodies.

shown to display a rounder, more cuboidal morphology, indicative of a more epithelial phenotype. Conversely, cells that engage ␣v integrins typically display a more elongated morphology, accompanied by aligned actin filaments, which is indicative of a more mesenchymal phenotype. Here we aimed to determine whether differential integrin engagement by RLE-6TN cells on the FNIII9⬘10 variants would elicit similar responses. Based on SPR and cell attachment results, cells should strongly engage ␣3 integrins on Ln and FNIII9⬘10 substrates, and therefore, these substrates would be expected to support a predominantly round, epithelial morphology. Indeed, cell circularity was highest on these substrates compared with all other conditions. Furthermore, inhibition of ␣3 integrin with antibodies resulted in a decrease in cell circularity on both Ln and FNIII9⬘10 (Fig. 10). Based on SPR and cell attachment results, cells should strongly engage ␣v integrin on FNIII9R-A10 and FnIII94G10, and therefore, these substrates would be expected to support a more elongated, mesenchymal phenotype. As expected, cell circularity was lowest on these substrates. Inhibition of ␣v integrin with antibodies resulted in an increase in cell circularity on both FNIII9R-A10 and FnIII94G10, whereas inhibition of ␣3 and ␣5 integrins had a minimal effect on cell circularity (Fig. 10).

Discussion These studies demonstrate that Fn binds integrin ␣3␤1 in a cell-free system, and this binding is dependent on the presence of the ninth type III repeat and its spacing relative to the tenth type III repeat, presumably mediated through the RGD and synergy sites. These results provide significant insight into the role of the central cell binding domain of Fn, i.e. the ninth and tenth type III repeats, and what is classically considered a Ln receptor, integrin ␣3␤1.

25544 JOURNAL OF BIOLOGICAL CHEMISTRY

The role of the PHSRN site in ␣5␤1 integrin binding to Fn is well characterized, and in addition, this site has been shown to enhance binding of additional integrins, as illustrated by its role in facilitating strong interactions between Fn and the platelet integrin ␣IIb␤3 (50). Studies suggest that the PHSRN site acts to stabilize the high affinity conformation of the RGD site required for ␣5␤1 integrin binding. Here we demonstrate that ␣3␤1 integrin binding to Fn is also dependent on the relative spacing of the RGD and PHSRN sites. Previous investigations to elucidate potential ligands for integrin ␣3␤1 have been conflicting regarding interactions with Fn. Many of these studies involve cell-based attachment assays, which are at times difficult to interpret, especially in the case of integrin ␣3␤1, in which binding affinity to ECM ligands is influenced by the presence of additional integrins (51). Studies utilizing soluble integrins in cell free systems are also conflicting. For example, studies by Eble et al. (28) reported minimal ␣3␤1 binding to Fn. Interestingly, these studies showing minimal binding were performed on tissue culture plastic, whereas several studies confirming Fn/␣3␤1 interactions utilized ECM affinity columns (29, 52). Our SPR studies possibly bridge these discrepancies. Fn has been shown to undergo unfolding upon adsorption as a consequence of the hydrophobic effect and the protein adopting its lowest energy state (53–56). Such unfolding events likely disrupt the relative positioning of the RGD and PHSRN sites and, according to the SPR studies presented here, would disrupt ␣3␤1 binding. This conclusion is further supported by data demonstrating that fulllength Fn in solution binds ␣3␤1 exceptionally well, as indicated by the presented SPR results. Because ␣3␤1 and ␣5␤1 integrin are involved in epithelial wound repair, these findings that ␣3␤1 integrin, like ␣5␤1 integrin, binds to Fn in a synergy-dependent manner have implications in cell interactions during epithelial wound healVOLUME 290 • NUMBER 42 • OCTOBER 16, 2015

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

FIGURE 10. Cell circularity on FnIII9ⴕ10 variants in the presence of integrin blocking antibodies. To determine the influence of integrin engagement on RLE-6TN cell spreading responses on FnIII9⬘10 variants, RLE-6TN cells were incubated with ␣3, ␣5, or ␣v antibodies for 30 min prior to plating on FnIII9⬘10, FnIII9R-A10, FnIII92G10, FnIII94G10, Fn, and Ln. Cumulative frequency distribution of cell circularity was utilized to visualize shifts in the distribution of cell circularity on these substrates in the presence of integrin blocking antibodies.

ing. Alveolar epithelial type II cells increase expression of ␣5␤1 integrin in response to injury (22); therefore, it is likely that integrin ␣3␤1 binds Fn with great affinity in this cell type in early wound repair before ␣5␤1 integrin is highly expressed. Although ␣5␤1 appears to be the main Fn receptor during alveolar wound repair, integrin blocking experiments with airway epithelial cells showed that in addition to ␣5, ␤1, and Fn blocking antibodies, ␣3 blocking antibodies also resulted in a significant decrease in wound repair (22). Furthermore, it has been shown that integrin ␣3␤1 binding to Fn can be affected by the presence of ␣5␤1 integrin such that ␣3␤1 binding is low in cells expressing ␣3␤1 and ␣5␤1 at comparable levels, but ␣3␤1 binding to Fn is greatly enhanced in H69 cells that highly express ␣3␤1 compared with other integrins (29). These reports, taken along with our findings here, suggest that ␣3␤1 binding to Fn could potentially modulate wound repair by facilitating binding to ECM in a wound healing environment as the ECM composition transitions from predominantly Ln to high levels of Fn. Control SPR experiments demonstrate similar affinities of all FnIII9⬘10 variants for integrin ␣v␤3, with all FnIII9⬘10 variants showing affinities in the range of 30 – 65 nM. The RGD and PHSRN sites have previously been described as a possible on/off switch for ␣5␤1 binding to Fn (25). Our findings show that in the context of epithelial cells, destabilizing the PHSRN and RGD sites may additionally serve as an off switch for ␣3␤1/ ␣5␤1 binding and on for ␣v integrin binding. Highlighting the significance of such a possible integrin switch, binding of ␣3␤1/ ␣5␤1 integrins have markedly different effects on epithelial phenotype compared with ␣v integrin binding. Integrin ␣3␤1 epithelial cell interactions are associated with maintenance of epithelial phenotype and wound healing, whereas interactions OCTOBER 16, 2015 • VOLUME 290 • NUMBER 42

with ␣v integrins have been associated with aberrant wound healing, scar tissue formation, epithelial to mesenchymal transitions, and tumor metastasis (22, 27, 34, 36). The extracellular microenvironment of disease states associated with increased ␣v integrin binding, scar tissue in particular, is associated with contractile fibroblasts (3). Evidence suggests that Fn is capable of undergoing force-mediated unfolding in response to cellular forces (48, 50), which has been hypothesized to lead to an increase in spacing between the RGD and PHSRN sites within the integrin binding domain. Our data suggest that if such a speculative force-activated conformational change does occur, this would lead to diminished integrin binding to Fn while not affecting ␣v binding, which could further contribute to the nonhomeostatic condition. These studies provide insights into how Fn interactions could potentially contribute to normal versus pathological wound healing. Author Contributions—A. C. B. and T. H. B. designed the studies and wrote the paper. A. C. B. designed, performed, and analyzed SPR and cell spreading experiments. M. M. D. performed and analyzed cell attachment experiments. K. C. C. designed and performed CD experiments. S. E. S. provided technical assistance and contributed to SPR analysis. All authors reviewed the results and approved the final version of the manuscript.

References 1. Humphries, J. D., Byron, A., and Humphries, M. J. (2006) Integrin ligands at a glance. J. Cell Sci. 119, 3901–3903 2. Markowski, M. C., Brown, A. C., and Barker, T. H. (2012) Directing epithelial to mesenchymal transition through engineered microenvironments displaying orthogonal adhesive and mechanical cues. J. Biomed.

JOURNAL OF BIOLOGICAL CHEMISTRY

25545

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9 Mater. Res. A 100, 2119 –2127 3. Brown, A. C., Fiore, V. F., Sulchek, T. A., and Barker, T. H. (2013) Physical and chemical microenvironmental cues orthogonally control the degree and duration of fibrosis-associated epithelial-to-mesenchymal transitions. J. Pathol. 229, 25–35 4. Clause, K. C., and Barker, T. H. (2013) Extracellular matrix signaling in morphogenesis and repair. Curr. Opin. Biotechnol. 24, 830 – 833 5. Ruoslahti, E. (1996) RGD and other recognition sequences for integrins. Annu. Rev. Cell Dev. Biol. 12, 697–715 6. Humphries, M. J. (1990) The molecular basis and specificity of integrinligand interactions. J. Cell Sci. 97, 585–592 7. Plow, E. F., Haas, T. A., Zhang, L., Loftus, J., and Smith, J. W. (2000) Ligand binding to integrins. J. Biol. Chem. 275, 21785–21788 8. Mao, Y., and Schwarzbauer, J. E. (2005) Fibronectin fibrillogenesis, a cellmediated matrix assembly process. Matrix Biol. 24, 389 –399 9. Pankov, R., and Yamada, K. (2002) Fibronectin at a glance. J. Cell Sci. 115, 3861–3863 10. Baneyx, G., Baugh, L., and Vogel, V. (2002) Fibronectin extension and unfolding within cell matrix fibrils controlled by cytoskeletal tension. Proc. Natl. Acad. Sci. U.S.A. 99, 5139 –5143 11. Barker, T. H., Baneyx, G., Cardó-Vila, M., Workman, G. A., Weaver, M., Menon, P. M., Dedhar, S., Rempel, S. A., Arap, W., Pasqualini, R., Vogel, V., and Sage, E. H. (2005) SPARC regulates extracellular matrix organization through its modulation of integrin-linked kinase activity. J. Biol. Chem. 280, 36483–36493 12. Erickson, H. P. (1994) Reversible unfolding of fibronectin type III and immunoglobulin domains provides the structural basis for stretch and elasticity of titin and fibronectin. Proc. Natl. Acad. Sci. U.S.A. 91, 10114 –10118 13. Mardon, H. J., and Grant, K. E. (1994) The role of the ninth and tenth type III domains of human fibronectin in cell adhesion. FEBS Lett. 340, 197–201 14. Mould, A. P., Askari, J. A., Aota, S., Yamada, K. M., Irie, A., Takada, Y., Mardon, H. J., and Humphries, M. J. (1997) Defining the topology of integrin ␣5␤1-fibronectin interactions using inhibitory anti-␣5 and anti-␣1 monoclonal antibodies. Evidence that the synergy sequence of fibronectin is recognized by the amino-terminal repeats of the ␣5 subunit. J. Biol. Chem. 272, 17283–17292 15. Aota, S., Nomizu, M., and Yamada, K. M. (1994) The short amino acid sequence Pro-His-Ser-Arg-Asn in human fibronectin enhances cell-adhesive function. J. Biol. Chem. 269, 24756 –24761 16. Krammer, A., Craig, D., Thomas, W. E., Schulten, K., and Vogel, V. (2002) A structural model for force regulated integrin binding to fibronectin’s RGD-synergy site. Matrix Biol 21, 139 –147 17. Grant, R. P., Spitzfaden, C., Altroff, H., Campbell, I. D., and Mardon, H. J. (1997) Structural requirements for biological activity of the ninth and tenth FIII domains of human fibronectin. J. Biol. Chem. 272, 6159 – 6166 18. Altroff, H., Schlinkert, R., van der Walle, C. F., Bernini, A., Campbell, I. D., Werner, J. M., and Mardon, H. J. (2004) Interdomain tilt angle determines integrin-dependent function of the ninth and tenth FIII domains of human fibronectin. J. Biol. Chem. 279, 55995–56003 19. Altroff, H., van der Walle, C. F., Asselin, J., Fairless, R., Campbell, I. D., and Mardon, H. J. (2001) The eighth FIII domain of human fibronectin promotes integrin ␣5␤1 binding via stabilization of the ninth FIII domain. J. Biol. Chem. 276, 38885–38892 20. Redick, S. D., Settles, D. L., Briscoe, G., and Erickson, H. P. (2000) Defining fibronectin’s cell adhesion synergy site by site-directed mutagenesis. J. Cell Biol. 149, 521–527 21. Chada, D., Mather, T., and Nollert, M. U. (2006) The synergy site of fibronectin is required for strong interaction with the platelet integrin ␣IIb␤3. Ann. Biomed. Eng. 34, 1542–1552 22. Hérard, A. L., Pierrot, D., Hinnrasky, J., Kaplan, H., Sheppard, D., Puchelle, E., and Zahm, J. M. (1996) Fibronectin and its ␣5␤1-integrin receptor are involved in the wound-repair process of airway epithelium. Am. J. Physiol. 271, L726 –L733 23. Martino, M. M., Mochizuki, M., Rothenfluh, D. A., Rempel, S. A., Hubbell, J. A., and Barker, T. H. (2009) Controlling integrin specificity and stem cell differentiation in 2D and 3D environments through regulation of fi-

25546 JOURNAL OF BIOLOGICAL CHEMISTRY

bronectin domain stability. Biomaterials 30, 1089 –1097 24. Khew, S. T., Zhu, X. H., and Tong, Y. W. (2007) An integrin-specific collagen-mimetic peptide approach for optimizing Hep3B liver cell adhesion, proliferation, and cellular functions. Tissue Eng. 13, 2451–2463 25. Carson, A. E., and Barker, T. H. (2009) Emerging concepts in engineering extracellular matrix variants for directing cell phenotype. Regen. Med. 4, 593– 600 26. Brown, A. C., Rowe, J. A., and Barker, T. H. (2011) Guiding epithelial cell phenotypes with engineered integrin-specific recombinant fibronectin fragments. Tissue Eng. Part A 17, 139 –150 27. Sheppard, D. (2003) Functions of pulmonary epithelial integrins: from development to disease. Physiol. Rev. 83, 673– 686 28. Eble, J. A., Wucherpfennig, K. W., Gauthier, L., Dersch, P., Krukonis, E., Isberg, R. R., and Hemler, M. E. (1998) Recombinant soluble human ␣3␤1 integrin: purification, processing, regulation, and specific binding to laminin-5 and invasin in a mutually exclusive manner. Biochemistry 37, 10945–10955 29. Elices, M. J., Urry, L. A., and Hemler, M. E. (1991) Receptor functions for the integrin VLA-3: fibronectin, collagen, and laminin binding are differentially influenced by Arg-Gly-Asp peptide and by divalent cations. J. Cell Biol. 112, 169 –181 30. Kaufmann, R., Frösch, D., Westphal, C., Weber, L., and Klein, C. E. (1989) Integrin VLA-3: ultrastructural localization at cell-cell contact sites of human cell cultures. J. Cell Biol. 109, 1807–1815 31. Sriramarao, P., Steffner, P., and Gehlsen, K. R. (1993) Biochemical evidence for a homophilic interaction of the ␣3␤1 integrin. J. Biol. Chem. 268, 22036 –22041 32. Kim, K. K., Wei, Y., Szekeres, C., Kugler, M. C., Wolters, P. J., Hill, M. L., Frank, J. A., Brumwell, A. N., Wheeler, S. E., Kreidberg, J. A., and Chapman, H. A. (2009) Epithelial cell ␣3␤1 integrin links ␤-catenin and Smad signaling to promote myofibroblast formation and pulmonary fibrosis. J. Clin. Invest. 119, 213–224 33. Kim, Y., Kugler, M. C., Wei, Y., Kim, K. K., Li, X., Brumwell, A. N., and Chapman, H. A. (2009) Integrin ␣3␤1-dependent ␤-catenin phosphorylation links epithelial Smad signaling to cell contacts. J. Cell Biol. 184, 309 –322 34. Lubman, R. L., Zhang, X. L., Zheng, J., Ocampo, L., Lopez, M. Z., Veeraraghavan, S., Zabski, S. M., Danto, S. I., and Borok, Z. (2000) Integrin ␣3-subunit expression modulates alveolar epithelial cell monolayer formation. Am. J. Physiol. Lung Cell Mol. Physiol. 279, L183–L193 35. Kreidberg, J. A., Donovan, M. J., Goldstein, S. L., Rennke, H., Shepherd, K., Jones, R. C., and Jaenisch, R. (1996) ␣3␤1 integrin has a crucial role in kidney and lung organogenesis. Development 122, 3537–3547 36. Kim, K. K., Kugler, M. C., Wolters, P. J., Robillard, L., Galvez, M. G., Brumwell, A. N., Sheppard, D., and Chapman, H. A. (2006) Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc. Natl. Acad. Sci. U.S.A. 103, 13180 –13185 37. Stabenfeldt, S. E., Gossett, J. J., and Barker, T. H. (2010) Building better fibrin knob mimics: an investigation of synthetic fibrin knob peptide structures in solution and their dynamic binding with fibrinogen/fibrin holes. Blood 116, 1352–1359 38. Petrie, T. A., Capadona, J. R., Reyes, C. D., and García, A. J. (2006) Integrin specificity and enhanced cellular activities associated with surfaces presenting a recombinant fibronectin fragment compared to RGD supports. Biomaterials 27, 5459 –5470 39. Morton, T. A., and Myszka, D. G. (1998) Kinetic analysis of macromolecular interactions using surface plasmon resonance biosensors. Methods Enzymol. 295, 268 –294 40. Joss, L., Morton, T. A., Doyle, M. L., and Myszka, D. G. (1998) Interpreting kinetic rate constants from optical biosensor data recorded on a decaying surface. Anal. Biochem. 261, 203–210 41. Myszka, D. G., and Morton, T. A. (1998) CLAMP: a biosensor kinetic data analysis program. Trends Biochem. Sci. 23, 149 –150 42. Koteliansky, V. E., Glukhova, M. A., Bejanian, M. V., Smirnov, V. N., Filimonov, V. V., Zalite, O. M., and Venyaminov, S. (1981) A study of the structure of fibronectin. Eur. J. Biochem. 119, 619 – 624 43. Litvinovich, S. V., Brew, S. A., Aota, S., Akiyama, S. K., Haudenschild, C.,

VOLUME 290 • NUMBER 42 • OCTOBER 16, 2015

Integrin ␣3␤1-Fibronectin Binding Dependent on FnIII9

44.

45.

46.

47.

48.

49.

and Ingham, K. C. (1998) Formation of amyloid-like fibrils by self-association of a partially unfolded fibronectin type III module. J. Mol. Biol. 280, 245–258 Pereira, P., Kelly, S. M., Gellert, P. R., and van der Walle, C. F. (2008) Interdomain mobility and conformational stability of type III fibronectin domain pairs control surface adsorption, desorption and unfolding. Colloids Surfaces B Biointerfaces 64, 1–9 Mould, A. P., Akiyama, S. K., and Humphries, M. J. (1995) Regulation of integrin ␣5␤1-fibronectin interactions by divalent cations. Evidence for distinct classes of binding sites for Mn2⫹, Mg2⫹, and Ca2⫹. J. Biol. Chem. 270, 26270 –26277 Danen, E. H., Aota, S., van Kraats, A. A., Yamada, K. M., Ruiter, D. J., and van Muijen, G. N. (1995) Requirement for the synergy site for cell adhesion to fibronectin depends on the activation state of integrin ␣5␤1. J. Biol. Chem. 270, 21612–21618 Rao, S. P., Gehlsen, K. R., and Catanzaro, A. (1992) Identification of a ␤1 integrin on Mycobacterium avium-Mycobacterium intracellulare. Infect. Immun. 60, 3652–3657 Smith, M. L., Gourdon, D., Little, W. C., Kubow, K. E., Eguiluz, R. A., Luna-Morris, S., and Vogel, V. (2007) Force-induced unfolding of fibronectin in the extracellular matrix of living cells. PLoS Biol. 5, e268 Bachan, P., Kim, H. J., and Ingbar, D. H. (2009) Alveolar epithelial repair following acute lung injury. In C68: Alveolar Epithelium, pp. A4996, American Thoracic Society, New York

OCTOBER 16, 2015 • VOLUME 290 • NUMBER 42

50. Cao, L., Zeller, M. K., Fiore, V. F., Strane, P., Bermudez, H., and Barker, T. H. (2012) Phage-based molecular probes that discriminate force-induced structural states of fibronectin in vivo. Proc. Natl. Acad. Sci. U.S.A. 109, 7251–7256 51. Hodivala-Dilke, K. M., DiPersio, C. M., Kreidberg, J. A., and Hynes, R. O. (1998) Novel roles for ␣3␤1 integrin as a regulator of cytoskeletal assembly and as a trans-dominant inhibitor of integrin receptor function in mouse keratinocytes. J. Cell Biol. 142, 1357–1369 52. Elices, M. J., and Hemler, M. E. (1989) The human integrin VLA-2 is a collagen receptor on some cells and a collagen/laminin receptor on others. Proc. Natl. Acad. Sci. U.S.A. 86, 9906 –9910 53. Underwood, P. A., Steele, J. G., and Dalton, B. A. (1993) Effects of polystyrene surface chemistry on the biological activity of solid phase fibronectin and vitronectin, analysed with monoclonal antibodies. J. Cell Sci. 104, 793– 803 54. Pitt, W. G., Weaver, D. R., and Cooper, S. L. (1993) Fibronectin adsorpton kinetics on phase segregated polyurethaneureas. J. Biomater. Sci. Polym. Ed. 4, 337–346 55. Narasimhan, C., and Lai, C. S. (1989) Conformational changes of plasma fibronectin detected upon adsorption to solid substrates: a spin-label study. Biochemistry 28, 5041–5046 56. Wolff, C., and Lai, C. S. (1989) Fluorescence energy transfer detects changes in fibronectin structure upon surface binding. Arch. Biochem. Biophys. 268, 536 –545

JOURNAL OF BIOLOGICAL CHEMISTRY

25547

Integrin α3β1 Binding to Fibronectin Is Dependent on the Ninth Type III Repeat.

Fibronectin (Fn) is a promiscuous ligand for numerous cell adhesion receptors or integrins. The vast majority of Fn-integrin interactions are mediated...
NAN Sizes 1 Downloads 7 Views