Study of the interaction between sialic acid-binding immunoglobulin-type lectins (Siglec) and sialylated glycans for the development of a new generation of immunomodulators.

Abstract

Glycans and complementary glycan-binding proteins represent essential components in the control of both innate and adaptive immunity. Sialic acids are a family of sugars found on the terminal end of mammalian glycoconjugates; they able to act as marker of self in the immune system, as such residues are absent in most microbes. Sialic acid-binding immunoglobulin-like lectins, or Siglecs, are cell surface receptors that recognize sialic acids and are known to modulate immune responses, influencing almost every cell in the hematopoietic system. Siglecs are involved in events like cell adhesion and signaling, inhibition or regulation of the immune cell activation, all mediated by the interaction with sialylated ligands. Sialic acid-Siglec interactions have been associated with a broad spectrum of diseases, stretching from autoimmunity to neurodegeneration and cancer. Thus, strategies for a rational modulation of the interactions between Siglecs and sialylated glycans in pathophysiological processes exhibit a great therapeutic potential. In this context, the present thesis project aimed at the study of the interaction between Siglecs and their cognate sialic acid containing ligands, to disclose the key recognition events underlining host immune suppression or activation. To this end, a multidisciplinary approach combining advanced technologies as ligand-based NMR techniques, including STD-NMR and tr-NOESY, biophysical binding assays and computational methodologies, such as homology modelling docking and MD simulations, was applied to provide an atomistic depiction of the interaction interfaces between various sialoglycans and their receptors. The described strategy has been employed to characterize the binding features of several receptors of the Siglecs family, namely CD22/Siglec-2, Siglec-10 and Siglec-7. CD22 is a B-cell surface inhibitory protein capable of selectively -(2,6) linked sialylated glycans, thus dampening autoimmune responses against self-antigens. The characterization of complex-type N-glycans by CD22 allowed to describe the conformational behavior of the flexible ligands; the formation of CD22 homo-oligomers on the B-cell surface was also addressed. Furthermore, it was provided a global vision of how the most diffuse neuraminic acid forms of sialylated N-glycans are accomodated in the binding pocket of CD22. Moreover, the elucidation of the binding epitope of a synthetic sialo-mimetic upon CD22 interaction afforded new hints for the design and synthesis of high-affinity ligands of therapeutic relevance against B-cell derived malignancies. Then, the Siglec-10, an inhibitory receptor that recognize 2,3 and -linked sialoglycans was studied, thus providing the first insights of the molecular mechanisms regulating the interaction between Siglec-10 and naturally occurring sialoglycans. After that, Siglec-7, a well-established inhibitory receptor that is primarily located on natural killer where it acts as inhibitor of cancer cells cytotoxicity via sialylated ligands binding, has been characterized in the interplay with the oncogenic pathogen F. nucleatum. Indeed, the presence of sialylated lipopolysaccharide (LPS) on certain F. nucleatum strains, hinted that it may have a significant role at the immune interface. The interaction between Siglec-7 and the O-polysaccharide chain from the LPS of F. nucleatum 10953 strain has been depicted, thus delineating a structural binding model that might contribute to explain the etiological role of F. nucleatum in carcinogenesis. A similar approach was employed to other sialoglycan- related systems, i. e. to dissect the mechanism of sialic acid recognition and hydrolysis by mumps virus hemagglutinin neuraminidase, a viral glycoprotein that plays key roles in virus entry and infection; and to assess the binding of the human macrophage galactose-type lectin (MGL) in the interplay with lipooligosaccharide of E. coli strain R1. In conclusion, the structural and functional characterization of Siglec- sialylated glycans interaction have allowed the analysis, at a molecular level, of crucial feature of 3D complexes, highlighting the molecular determinants involved in recognition and binding events, that will aid for the development or optimization of molecules for therapeutic targeting of the Siglecs

    Similar works