4.8 Article

Hedgehog Signaling Regulates Metabolism and Polarization of Mammary Tumor-Associated Macrophages

Journal

CANCER RESEARCH
Volume 81, Issue 21, Pages 5425-5437

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/0008-5472.CAN-20-1723

Keywords

-

Categories

Funding

  1. Department of Defense [W81XWH-14-1-0516, W81XWH-18-1-0036, W81XWH-10-1-0755]
  2. NCI [R01CA169202]
  3. Breast Cancer Research Foundation of Alabama (BCRFA)
  4. VA [BXAl3374]
  5. NCI/NIH [CA194048]
  6. NIH [P30 AR048311, P30 AI027667]
  7. UAB Comprehensive Cancer Center's Preclinical Imaging Shared Facility - NIH [P30 CA013148]

Ask authors/readers for more resources

The oncogenic Hedgehog (Hh) signaling plays a novel role in programming signature metabolic circuitries that regulate the alternative polarization of tumor-associated macrophages. Inhibiting Hh signaling induces significant metabolic alterations in macrophages and shifts their metabolism from fatty acid oxidation to glycolysis, promoting immune-suppressive polarization.
Elevated infiltration of immunosuppressive alternatively polarized (M2) macrophages is associated with poor prognosis in patients with cancer. The tumor microenvironment remarkably orchestrates molecular mechanisms that program these macrophages. Here we identify a novel role for oncogenic Hedgehog (Hh) signaling in programming signature metabolic circuitries that regulate alternative polarization of tumor-associated macrophages. Two immunocompetent orthotopic mouse models of mammary tumors were used to test the effect of inhibiting Hh signaling on tumor-associated macrophages. Treatment with the pharmacologic Hh inhibitor vismodegib induced a significant shift in the profile of tumor-infiltrating macrophages. Mass spectrometry-based metabolomic analysis showed Hh inhibition induced significant alterations in metabolic processes, including metabolic sensing, mitochondrial adaptations, and lipid metabolism. In particular, inhibition of Hh in M2 macrophages reduced flux through the UDP-GlcNAc biosynthesis pathway. Consequently, O-GlcNAc-modification of STAT6 decreased, mitigating the immune-suppressive program of M2 macrophages, and the metabolically demanding M2 macrophages shifted their metabolism and bioenergetics from fatty acid oxidation to glycolysis. M2 macrophages enriched from vismode-gib-treated mammary tumors showed characteristically decreased O-GlcNAcylation and altered mitochondrial dynamics. These Hh-inhibited macrophages are reminiscent of inflammatory (M1) macrophages, phenotypically characterized by fragmented mitochondria. This is the first report highlighting the relevance of Hh signaling in controlling a complex metabolic network in immune cells. These data describe a novel immunometabolic function of Hh signaling that can be clinically exploited. Significance: These findings illustrate that Hh activity regulates a metabolic and bioenergetic regulatory program in tumor-associated macrophages that promotes their immune-suppressive polarization.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available