Hedgehog (HH) signaling acts throughout embryonic development to ensure proper tissue patterning and organ formation, while in adults HH signaling is vital for tissue maintenance and homeostasis. Aberrant HH pathway activity contributes to numerous developmental diseases and drives the initiation and progression of several cancers, including basal cell carcinoma, rhabdomyosarcoma, and medulloblastoma. More recently, de-regulated HH signaling has been implicated in multiple additional cancers, including pancreatic cancer, where tumor cell-derived HH ligands signal in a paracrine manner to the surrounding microenvironment. In particular, pancreatic cancer is characterized by an extensive HH-responsive fibrotic stroma that promotes tumor growth. However, strikingly little is known about the mechanisms by which HH signaling acts in pancreatic cancer. We propose to address this critical gap in our knowledge through a systematic analysis of the requirement for dosage-dependent HH signaling in pancreatic tumor growth. Through a collaborative effort, the Allen and the Pasca di Magliano laboratories have determined that altering the dosage of HH signaling through targeting of the HH co-receptors GAS1, BOC and CDON differentially regulates pancreatic tumor growth. Surprisingly, pancreatic fibroblasts lacking two of these co-receptors (GAS1 and BOC) have increased tumor-promoting ability compared to wild-type pancreatic fibroblasts when co-injected with tumor cells in mice. In contrast, pancreatic fibroblasts lacking all three co-receptors are unable to promote pancreatic tumor growth. These data suggest that the dosage of HH signaling dramatically affects pancreatic tumor growth. Further, these findings have important clinical implications, and provide an explanation for the negative outcome of recent clinical trials of HH pathway inhibitors in pancreatic cancer. Thus, the central question in this proposal is: how do different levels of HH pathway activity affect pancreatic tumor growth? This proposal continues a unique collaboration between the Allen and Pasca di Magliano labs that combines expertise in HH signaling and pancreatic cancer to test the hypothesis that the level of HH pathway activity is an essential regulator of pancreatic tumor growth. Thus, the objective of this proposal is to define the role of dosage-dependent HH signaling in pancreatic cancer through the dissection of the requirement for 1) HH ligands, 2) HH receptors and 3) downstream HH signal transduction components in pancreatic tumor growth. The expected outcomes of this work include: 1) a comprehensive analysis of the requirement for different levels of tumor-derived HH ligands in pancreatic tumor growth, 2) the elucidation of the relative contributions of cell surface HH receptors and antagonists to stromal promotion of pancreatic cancer, and 3) a determination of the effects of altered downstream HH pathway activity on tumor growth and metastasis. We expect that these results will define the role of HH signaling in pancreatic tumor growth and will provide insight into novel approaches to successfully target the HH pathway in the treatment of pancreatic cancer and other HH-driven pathologies.

Public Health Relevance

Aberrant HH signaling drives the initiation and progression of a growing number of cancers; however, surprisingly little is known about the mechanisms by which HH signaling contributes to tumorigenesis. This proposal will address an unexpected role for dosage-dependent HH signaling in pancreatic cancer, the fourth leading cause of cancer-related death in the United States, with a five-year survival rate of less than five percent. The studies proposed here will identify novel approaches to target HH signaling in the treatment of pancreatic cancer, which currently lacks effective treatments, and other HH-dependent pathologies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA198074-04
Application #
9544105
Study Section
Tumor Microenvironment Study Section (TME)
Program Officer
Snyderwine, Elizabeth G
Project Start
2015-09-25
Project End
2020-08-31
Budget Start
2018-09-01
Budget End
2019-08-31
Support Year
4
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Michigan Ann Arbor
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Pal, Anupama; Dziubinski, Michele; Di Magliano, Marina Pasca et al. (2018) Usp9x Promotes Survival in Human Pancreatic Cancer and Its Inhibition Suppresses Pancreatic Ductal Adenocarcinoma In Vivo Tumor Growth. Neoplasia 20:152-164
Schofield, Heather K; Zeller, Jörg; Espinoza, Carlos et al. (2018) Mutant p53R270H drives altered metabolism and increased invasion in pancreatic ductal adenocarcinoma. JCI Insight 3:
Schofield, Heather K; Tandon, Manuj; Park, Min-Jung et al. (2018) Pancreatic HIF2? Stabilization Leads to Chronic Pancreatitis and Predisposes to Mucinous Cystic Neoplasm. Cell Mol Gastroenterol Hepatol 5:169-185.e2
Zhang, Yaqing; Velez-Delgado, Ashley; Mathew, Esha et al. (2017) Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 66:124-136
Zhang, Yaqing; Yan, Wei; Mathew, Esha et al. (2017) Epithelial-Myeloid cell crosstalk regulates acinar cell plasticity and pancreatic remodeling in mice. Elife 6:
Hong, Mingi; Srivastava, Kshitij; Kim, Sungjin et al. (2017) BOC is a modifier gene in holoprosencephaly. Hum Mutat 38:1464-1470
Pinskey, Justine M; Franks, Nicole E; McMellen, Alexandra N et al. (2017) Neuropilin-1 promotes Hedgehog signaling through a novel cytoplasmic motif. J Biol Chem 292:15192-15204
Kumari, Archana; Ermilov, Alexandre N; Grachtchouk, Marina et al. (2017) Recovery of taste organs and sensory function after severe loss from Hedgehog/Smoothened inhibition with cancer drug sonidegib. Proc Natl Acad Sci U S A 114:E10369-E10378
Syu, Li-Jyun; Zhao, Xinyi; Zhang, Yaqing et al. (2016) Invasive mouse gastric adenocarcinomas arising from Lgr5+ stem cells are dependent on crosstalk between the Hedgehog/GLI2 and mTOR pathways. Oncotarget 7:10255-70
Yang, Shenghong; Imamura, Yu; Jenkins, Russell W et al. (2016) Autophagy Inhibition Dysregulates TBK1 Signaling and Promotes Pancreatic Inflammation. Cancer Immunol Res 4:520-30

Showing the most recent 10 out of 17 publications