Abstract

We previously demonstrated that Arg-Gly-Asp (RGD)-containing ligand-mimetic inhibitors of integrins are unable to dissociate pre-formed integrin-fibronectin complexes (IFCs). These observations suggested that amino acid residues involved in integrin-fibronectin binding become obscured in the ligand-occupied state. Because the epitopes of some function-blocking anti-integrin monoclonal antibodies (mAbs) lie near the ligand-binding pocket, it follows that the epitopes of these mAbs may become shielded in the ligand-occupied state. Here, we tested whether function-blocking mAbs directed against α5β1 can interact with the integrin after it forms a complex with an RGD-containing fragment of fibronectin. We showed that the anti-α5 subunit mAbs JBS5, SNAKA52, 16, and P1D6 failed to disrupt IFCs and hence appeared unable to bind to the ligand-occupied state. In contrast, the allosteric anti-β1 subunit mAbs 13, 4B4, and AIIB2 could dissociate IFCs and therefore were able to interact with the ligand-bound state. However, another class of function-blocking anti-β1 mAbs, exemplified by Lia1/2, could not disrupt IFCs. This second class of mAbs was also distinguished from 13, 4B4, and AIIB2 by their ability to induce homotypic cell aggregation. Although the epitope of Lia1/2 was closely overlapping with those of 13, 4B4, and AIIB2, it appeared to lie closer to the ligand-binding pocket. A new model of the α5β1-fibronectin complex supports our hypothesis that the epitopes of mAbs that fail to bind to the ligand-occupied state lie within, or very close to, the integrin-fibronectin interface. Importantly, our findings imply that the efficacy of some therapeutic anti-integrin mAbs could be limited by epitope masking.

Highlights

  • Integrins are a large family of ␣- and ␤-heterodimeric transmembrane receptors that mediate many cell-cell and cell-matrix interactions [1]

  • To test the ability of monoclonal antibodies (mAbs) to affect the stability of integrin-fibronectin complexes (IFCs), mAbs were injected during the dissociation phase as described previously [35]

  • Three possible outcomes would be anticipated as follows: (i) if mAbs were unable to bind to the complexes, there would be no effect on the dissociation rate; (ii) if mAbs could bind to and cause disruption of IFCs, there would be an observed increase in the dissociation rate; or (iii) if mAbs could bind to IFCs without causing disruption, there would be an increase in surface plasmon resonance (SPR) signal due to mAb binding

Read more

Summary

Results

Function-blocking Anti-␣5 mAbs Cannot Disrupt Pre-formed ␣5␤1-Fibronectin Complexes—For surface plasmon resonance (SPR) assays, we used the recombinant proteins ␣5␤1-Fc [38], the 50-kDa fragment of fibronectin (3FN6 –10, “50K”), and a control inactive mutant 50K-KGE in which the RGD sequence is converted to Lys-Gly-Glu (Fig. 1). The results (Fig. 3, cyan sensorgrams) showed that 13, 4B4, and AIIB2 caused a marked (ϳ4-fold) increase in the dissociation rate, whereas Lia1/2 did not affect the dissociation rate (Fig. 3 legend) When these mAbs were pre-mixed with the integrin before binding to the fibronectin fragment (Fig. 3, blue sensorgrams), 13, 4B4, and AIIB2 showed a partial (ϳ70%) inhibition of complex formation, whereas Lia1/2 caused a near-complete inhibition, comparable with that seen for the function-blocking anti-␣5 mAbs. 13, 4B4, and AIIB2 can bind to and disrupt IFCs; members of this group of mAbs appear to have an allosteric mode of action [35]. Non-function-blocking mAbs Can Bind to but Do Not Disrupt Pre-formed ␣5␤1-Fibronectin Complexes—we tested the effect of the non-function-blocking anti-␣5 subunit

Epitope location
Mouse IgG
Discussion
Experimental Procedures
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call