Induction of heparanase via IL-10 correlates with a high infiltration of CD163+ M2-type tumor-associated macrophages in inflammatory breast carcinomas

Matrix Biol Plus. 2020 Feb 29:6-7:100030. doi: 10.1016/j.mbplus.2020.100030. eCollection 2020 May.

Abstract

Inflammatory breast cancer (IBC) is the most aggressive and lethal form of breast cancer, characterized by a high infiltration of tumor-associated macrophages and poor prognosis. To identify new biomarkers and to elucidate the molecular mechanisms underlying IBC pathogenesis, we investigated the expression pattern of heparanase (HPSE) and its activator cathepsin L (CTSL). First, we quantitated the HPSE and CTSL mRNA levels in a cohort of breast cancer patients after curative surgery (20 IBC and 20-non-IBC). We discovered that both HPSE and CTSL mRNA levels were significantly induced in IBC tissue vis-à-vis non-IBC patients (p <0 .05 and p <0 .001, respectively). According to the molecular subtypes, HPSE mRNA levels were significantly higher in carcinoma tissues of triple negative (TN)-IBC as compared to TN-non-IBC (p <0 .05). Mechanistically, we discovered that pharmacological inhibition of HPSE activity resulted in a significant reduction of invasiveness in the IBC SUM149 cell line. Moreover, siRNA-mediated HPSE knockdown significantly downregulated the expression of the metastasis-related gene MMP2 and the cancer stem cell marker CD44. We also found that IBC tumors revealed robust heparanase immune-reactivity and CD163+ M2-type tumor-associated macrophages, with a positive correlation of both markers. Moreover, the secretome of axillary tributaries blood IBC CD14+ monocytes and the cytokine IL-10 significantly upregulated HPSE mRNA and protein expression in SUM149 cells. Intriguingly, massively elevated IL-10 mRNA expression with a trend of positive correlation with HPSE mRNA expression was detected in carcinoma tissue of IBC. Our findings highlight a possible role played by CD14+ monocytes and CD163+ M2-type tumor-associated macrophages in regulating HPSE expression possibly via IL-10. Overall, we suggest that heparanase, cathepsin L and CD14+ monocytes-derived IL-10 may play an important role in the pathogenesis of IBC and their targeting could have therapeutic implications.

Keywords: CD163+ M2-type tumor-associated macrophages; CTSL, cathepsin L; Cathepsin L; ECM, extracellular matrix; ER, estrogen receptor; FFPE, Formalin-Fixed Paraffin-Embedded; HER-2, human epidermal growth factor receptor-2; HPSE, heparanase; HSPGs, heparan sulfate proteoglycans; Heparanase; IBC, inflammatory breast cancer;; IL-10; IRB, Institutional Review Board; Inflammatory breast cancer; Invasion; MMP2, matrix metalloproteinase2; MTT, 3-(4,5-dimethyl-2-yl)-2,5-diphenyltetrazolium bromide; OGT 2115, 2-[4-[[3-(4-Bromophenyl)-1-oxo-2-propenyl]amino]-3-fluorophenyl]-5-benzoxazoleacetic acid; PR, progesterone receptor; TAMs, tumor-associated macrophages; TN, triple negative; TNF-α, tumor necrosis factor-α; Triple negative subtype; qPCR, quantitative real-time PCR; rh IL-10, recombinant human interleukin-10.