Abstract 2103: Hedgehog signaling regulates metabolism and polarization of mammary tumor-associated macrophages

Cancer Research(2022)

引用 0|浏览3
暂无评分
摘要
Abstract In the United States, a woman has a 12% chance of developing breast cancer, and current treatments offer little relief to patients diagnosed with metastatic disease. Tumorigenesis and successful establishment of metastases depend upon tumor cell interactions with the surrounding immune microenvironment. Elevated tumor infiltration of immunosuppressive (M2) macrophages correlates with poor prognosis of breast cancer patients. The tumor microenvironment remarkably orchestrates molecular mechanisms that program these macrophages toward the M2 phenotype. Also, metabolic programming is instrumental in orchestrating the polarization of macrophages to assume an M1 (tumor-eradicating) or an M2 (tumor-promoting) phenotype. Aberrant activation of Hedgehog (Hh) signaling in breast cancer cells enables them to survive, proliferate, and metastasize, thus making it a promising target for breast cancer treatment. Hh signaling also enables a crosstalk between breast cancer cells and cells in their milieu, thus contributing to M2 macrophage polarization. We used two immunocompetent orthotopic mouse models of mammary tumors to test the effect of inhibiting Hh signaling on tumor-associated macrophages, and discovered that treatment with the pharmacologic Hh inhibitor, Vismodegib, induced a significant shift in the profile of tumor-infiltrating macrophages. We hypothesized that Hh activity calibrates the metabolism in macrophages, leading to enhanced M2 phenotype and function within the tumor microenvironment. Using a mass spectrometry-enabled untargeted metabolomics approach, we identified that inhibiting Hh signaling reduces flux through the hexosamine biosynthetic pathway, resulting in reduced cellular O-GlcNAcylation in M2 macrophages. This impinges upon diminished STAT6 O-GlcNAcylation, which consequently decreases fatty acid oxidation and ultimately enacts a metabolic cascade including lipid utilization, cellular bioenergetics, and mitochondrial dynamics. As such, inhibiting Hh activity mitigates the metabolomic and bioenergetic underpinnings of the immunosuppressive program of M2 macrophages, resulting in macrophages that are functionally and phenotypically reminiscent of inflammatory, anti-tumor macrophages. In conclusion, we discovered a novel role for Hh signaling in promoting polarization of tumor-associated macrophages to the M2 type through recalibrating their metabolic circuitries, ultimately leading to diminished M2 phenotype and function within the tumor microenvironment. This is the first evidence highlighting the relevance of Hh signaling in controlling a complex metabolic network in immune cells. This knowledge will help us to better understand how to target and diminish the pro-tumorigenic functions of tumor-infiltrating macrophages. Citation Format: Dominique C. Hinshaw, Ann Hanna, Tshering Lama-Sherpa, Brandon Metge, Sarah C. Kammerud, Gloria A. Benavides, Atul Kumar, Heba A. Alsheikh, Mateus Mota, Dongquan Chen, Scott Ballinger, Jeffrey C. Rathmell, Selvarangan Ponnazhagan, Victor Darley-Usmar, Rajeev S. Samant, Lalita A. Shevde. Hedgehog signaling regulates metabolism and polarization of mammary tumor-associated macrophages [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2103.
更多
查看译文
关键词
macrophages,hedgehog,regulates metabolism,tumor-associated
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要