Suppression of integrin α3β1 by α9β1 in the epidermis controls the paracrine resolution of wound angiogenesis

2017 
Development of wound therapies is hindered by poor understanding of combinatorial integrin function in the epidermis. In this study, we generated mice with epidermis-specific deletion of α3β1, α9β1, or both integrins as well as keratinocyte lines expressing these integrin combinations. Consistent with proangiogenic roles for α3β1, α3-null keratinocytes showed reduced paracrine stimulation of endothelial cell migration and survival, and wounds of epidermis-specific α3 knockout mice displayed impaired angiogenesis. Interestingly, α9β1 in keratinocytes suppressed α3β1-mediated stimulation of endothelial cells, and wounds of epidermis-specific α9 knockout mice displayed delayed vascular normalization and reduced endothelial apoptosis, indicating that α9β1 cross-suppresses α3β1 proangiogenic functions. Moreover, α9β1 inhibited α3β1 signaling downstream of focal adhesion kinase (FAK) autoactivation at the point of Src-mediated phosphorylation of FAK Y861/Y925. Finally, α9β1 cross-suppressed many α3β1-dependent genes, including the gene that encodes MMP-9, which we implicated as a regulator of integrin-dependent cross talk to endothelial cells. Our findings identify a novel physiological context for combinatorial integrin signaling, laying the foundation for therapeutic strategies that manipulate α9β1 and/or α3β1 during wound healing.
    • Correction
    • Source
    • Cite
    • Save
    • Machine Reading By IdeaReader
    61
    References
    22
    Citations
    NaN
    KQI
    []