Suppression of breast cancer-associated bone loss with osteoblast proteomes via Hsp90ab1/moesin-mediated inhibition of TGFβ/FN1/CD44 signaling

Theranostics. 2022 Jan 1;12(2):929-943. doi: 10.7150/thno.66148. eCollection 2022.

Abstract

Background: Bone is a frequent site of metastases from breast cancer, but existing therapeutic options are not satisfactory. Although osteoblasts have active roles in cancer progression by assisting the vicious bone-destructive cycle, we employed a counterintuitive approach of activating pro-tumorigenic Wnt signaling and examined the paradoxical possibility of developing osteoblast-derived tumor-suppressive, bone-protective secretomes. Methods: Wnt signaling was activated by the overexpression of Lrp5 and β-catenin in osteoblasts as well as a pharmacological agent (BML284), and the therapeutic effects of their conditioned medium (CM) were evaluated using in vitro cell cultures, ex vivo breast cancer tissues, and a mouse model of osteolysis. To explore the unconventional regulatory mechanism of the action of Wnt-activated osteoblasts, whole-genome proteomics analysis was conducted, followed by immunoprecipitation and gain- and loss-of-function assays. Results: While osteoblasts did not present any innate tumor-suppressing ability, we observed that the overexpression of Lrp5 and β-catenin in Wnt signaling made their CM tumor-suppressive and bone-protective. The growth of breast cancer cells and tissues was inhibited by Lrp5-overexpressing CM (Lrp5 CM), which suppressed mammary tumors and tumor-driven bone destruction in a mouse model. Lrp5 CM also inhibited the differentiation and maturation of bone-resorbing osteoclasts by downregulating NFATc1 and cathepsin K. The overexpression of Lrp5 upregulated osteopontin that enriched Hsp90ab1 (Hsp90 beta) and moesin (MSN) in Lrp5 CM. Hsp90ab1 and MSN are atypical tumor-suppressing proteins since they are multi-tasking, moonlighting proteins that promote tumorigenesis in tumor cells. Importantly, Hsp90ab1 immuno-precipitated latent TGFβ and inactivated TGFβ, whereas MSN interacted with CD44, a cancer stem-cell marker, as well as fibronectin 1, an ECM protein. Furthermore, Hsp90ab1 and MSN downregulated KDM3A that demethylated histones, together with PDL1 that inhibited immune responses. Conclusion: In contrast to inducing tumor-enhancing secretomes and chemoresistance in general by inhibiting varying oncogenic pathways in chemotherapy, this study presented the unexpected outcome of generation tumor-suppressive secretomes by activating the pro-tumorigenic Wnt pathway. The results shed light on the contrasting role of oncogenic signaling in tumor cells and osteoblast-derived secretomes, suggesting a counterintuitive option for the treatment of breast cancer-associated bone metastasis.

Keywords: CD44; Hsp90ab1; Moesin; breast cancer; osteoblasts.

MeSH terms

  • Animals
  • Breast Neoplasms / complications*
  • Breast Neoplasms / metabolism
  • Cell Line, Tumor
  • Disease Models, Animal
  • Female
  • Fibronectins / antagonists & inhibitors
  • Fibronectins / metabolism
  • HSP90 Heat-Shock Proteins / metabolism*
  • Humans
  • Hyaluronan Receptors / antagonists & inhibitors
  • Hyaluronan Receptors / metabolism
  • Mammary Neoplasms, Experimental / complications
  • Mammary Neoplasms, Experimental / therapy
  • Mice
  • Microfilament Proteins / metabolism*
  • Osteoblasts / metabolism*
  • Osteoclasts / metabolism
  • Osteogenesis
  • Osteolysis / metabolism
  • Osteolysis / prevention & control*
  • Proteome / metabolism
  • Secretome
  • Transforming Growth Factor beta / antagonists & inhibitors
  • Transforming Growth Factor beta / metabolism
  • Tumor Suppressor Proteins / metabolism*
  • Wnt Signaling Pathway*

Substances

  • CD44 protein, human
  • Fibronectins
  • HSP90 Heat-Shock Proteins
  • HSP90AB1 protein, human
  • Hyaluronan Receptors
  • Microfilament Proteins
  • Proteome
  • Transforming Growth Factor beta
  • Tumor Suppressor Proteins
  • moesin