Transfusion-related acute lung injury (TRALI) is a syndrome of respiratory distress which develops within 6 hours of blood transfusion. It is the leading cause of transfusion-related deaths and the pathogenesis is complex and incompletely understood. In the majority of the cases, anti-leukocyte antibodies present in the transfused blood product, in combination with recipient predisposing risk-factors such as inflammation, are implicated to be responsible for the onset of TRALI. Unfortunately, no therapies are available for TRALI.

Osteopontin (OPN) is an extracellular matrix protein with multiple biological functions. OPN is involved in normal physiological processes, such as cell migration and adhesion, but has also been implicated in a wide range of disease states, including cancer, atherosclerosis, glomerulonephritis, and several chronic inflammatory diseases. Interestingly, OPN is upregulated at sites of inflammation and tissue remodeling. As inflammation is an important risk factor for TRALI development, and as neutrophils (PMNs) are known effector cells in the pathogenesis of TRALI which migrate and accumulate in the lungs during TRALI development, we investigated the potential contribution of OPN in the onset of antibody-mediated TRALI.

We utilized a previously established murine TRALI model (Kapur et al, Blood 2017, Blood Advances 2018) in which C57BL/6 mice were first primed with a low dose of lipopolysaccharide (LPS) and depleted of their CD4+ T cells in vivo followed by injection of anti-major histocompatibility complex (MHC) class I antibodies (clones 34-1-2s and AF6-88.5.5.3). The TRALI response was analyzed after 90 minutes by analysis of pulmonary edema (lung wet-to-dry weight ratios, W/Ds) and the levels of pulmonary neutrophils. Wildtype (WT) mice suffered from antibody-mediated TRALI compared to untreated naïve mice, as was shown by their significantly increased lung W/Ds (4.72 vs 4.50, respectively, P<0.0001). This also corresponded to significantly increased levels of pulmonary PMNs compared to untreated naïve mice (34% vs 5%, P<0.0001). In contrast, C57BL/6 OPN knock-out mice were resistant to antibody-mediated TRALI induction as they did not display any significant increase in lung W/Ds levels or pulmonary PMNs compared to untreated naïve OPN knock-out mice (lung W/Ds 4.83 vs 4.75, and pulmonary PMNs 18% vs 16%, respectively). Strikingly, administration of purified recombinant OPN during TRALI induction in C57BL/6 OPN knock-out mice, significantly induced a TRALI reaction (lung W/Ds 5.12 vs 4.75 as compared to untreated naïve OPN knock-out mice, P<0.05). Mechanistically, this TRALI inducing effect of OPN administration to OPN knock-out mice was associated with increased levels of pulmonary PMNs (38% vs 16%, as compared to untreated naïve OPN knock-out mice, P<0.0001). In vivo blocking of OPN in WT mice with an anti-OPN antibody demonstrated decreased lung W/Ds as compared to treatment with an isotype antibody (4.48 vs 4.69, respectively, P<0.05). The OPN blocking response during TRALI was associated with a decreased level of pulmonary PMN accumulation as compared to treatment with an isotype antibody (14% vs 34%, respectively, P<0.0001). As the PMN-chemoattractant macrophage inflammatory protein (MIP)-2 has previously been described to be upregulated in murine antibody-mediated TRALI, we investigated if the OPN-associated pulmonary PMN accumulation and TRALI induction could be related to the levels of MIP-2. We found that plasma MIP-2 levels were increased in mice that were infused with anti-MHC class I antibodies as compared to naïve controls, but that addition or blocking of OPN did not affect these MIP-2 levels. This indicates that the OPN-related PMN responses in TRALI are independent of plasma MIP-2 levels.

Collectively, these data indicate OPN as a novel pathogenic factor which enhances antibody-mediated murine TRALI through stimulation of PMN migration towards the lungs, independent of MIP-2. This may suggest that blocking OPN (using an anti-OPN antibody) may prevent TRALI by impairing pulmonary PMN accumulation and could be a therapeutic avenue to explore in combatting this serious adverse complication of blood transfusion.

Disclosures

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution