Page 7 - Read Online
P. 7

Mazur et al. Rare Dis Orphan Drugs J 2023;2:1  https://dx.doi.org/10.20517/rdodj.2022.12  Page 3 of 10

               ELANE  MUTATIONS LEADING TO NEUTROPENIA
               During the promyelocytic stage of granulopoiesis in the bone marrow, NE is synthesized as zymogen and
               post-translationally activated in the neutrophil precursors [Figure 1A]. The NE activation steps include N-
               terminal trimming by cathepsin C, which is required for NE protease activity. In addition, NE may be
                                                                  [1]
               modified at C-terminus by an as yet unidentified protease . The proteolytically-processed C-terminal is
               thought to serve as a binding site for adaptor protein complex 3 (AP3), implicated in the transport of NE to
               lysosome-like granules [1,13] .

               NE mainly localizes to the primary granules of the neutrophil precursors, where it remains stored in a
               catalytically active, ready-to-use form. Neutrophils that are released from the bone marrow into the
               circulation, followed by recruitment to peripheral tissues, rely on catalytically active NE for their full
               spectrum of antimicrobial, pro-inflammatory and tissue-remodeling functions . In addition to the
                                                                                      [14]
               localization of NE to the primary granules in the neutrophil precursors and mature neutrophils, the
               presence of this protease out of the granules, including in cytosol, at the cell surface and in the nucleus, has
               also been documented [15-17] . In mature, circulating neutrophils, transient release of NE from the granules is
               likely to be required for the physiological functions of these cells, such as generating and releasing
               neutrophil extracellular traps, or regulating neutrophil migration [15,17] . However, NE misplaced from the
               granules in the neutrophil precursors in the bone marrow is associated with neutrophil pathology, such as
               SCN (see below).

               ELANE-based neutropenia results from the gain-of-function of mutant NE rather than diminished levels of
               native protein which might also be a consequence of monoallelic mutations . This is supported by several
                                                                                [1]
               lines of research. For example, a blockade of NE expression leads to the restoration of normal
               granulopoiesis and neutrophil differentiation, regardless of the type of underlying genetic mutation .
                                                                                                  [18]

               The ELANE gene consists of 5 exons and 6 introns, and > 200 mutations, distributed over all exons as well
               as some noncoding regions, have been described in association with congenital neutropenia [3,19] . However,
               to link genetic ELANE variants with pathologic outcomes, mechanistic studies are required. Mouse models
               of ELANE-targeted neutropenia have failed to accurately phenocopy human neutropenia because basal
               granulopoiesis was not disrupted in mice upon expression of abnormal murine NE, targeted at a position
               orthologous to human pathogenic NE mutation. A lack of suitable experimental neutropenia models has
               hampered the testing of ELANE mutants in vivo in the context of this pathology . Human, murine or rat
                                                                                    [20]
               promyelocytic cell lines transduced with mutant ELANE, or patient neutrophil or patient-derived induced
               pluripotent stem cell (iPSC) lines, are typically used to unravel the biochemical and biological consequences
               of different ELANE mutations in myeloid progenitors. On the basis of these studies, several ELANE
               mutations have been identified to interfere with neutrophil maturation potential [Table 1].

               According to recent studies, a comprehensive gene editing screen in the human hematopoietic stem and
               progenitor cells (HSPCs) dissects the genetic bases of ELANE neutropenia pathogenicity. This model, in
               which HSPCs were edited with sgRNAs that targeted a broad range of ELANE mutants in vitro, faithfully
                                                                          [21]
               recapitulated the known genetic features of ELANE-based neutropenia . In addition, human-edited HSPCs
               have also been shown to produce an ELANE pathogenic variant-dependent abnormal hematopoietic
               engraftment function, following infusion into specific immunodeficient mice. Thus, these new xenograft
               models of gene-edited human HSPCs also enable the outcome of ELANE mutations to be tested in in vivo
               settings, which was not possible before with ELANE transgenic mice .
                                                                        [21]
   2   3   4   5   6   7   8   9   10   11   12