1
|
Sigafoos AN, Tolosa EJ, Carr RM, Fernandez-Barrena MG, Almada LL, Pease DR, Hogenson TL, Raja Arul GL, Mousavi F, Sen S, Vera RE, Marks DL, Flores LF, LaRue-Nolan KC, Wu C, Bamlet WR, Vrabel AM, Sicotte H, Schenk EL, Smyrk TC, Zhang L, Rabe KG, Oberg AL, Zaphiropoulos PG, Chevet E, Graham RP, Hagen CE, di Magliano MP, Elsawa SF, Pin CL, Mao J, McWilliams RR, Fernandez-Zapico ME. KRAS Promotes GLI2-Dependent Transcription during Pancreatic Carcinogenesis. CANCER RESEARCH COMMUNICATIONS 2024; 4:1677-1689. [PMID: 38896052 PMCID: PMC11232480 DOI: 10.1158/2767-9764.crc-23-0464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/19/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
Aberrant activation of GLI transcription factors has been implicated in the pathogenesis of different tumor types including pancreatic ductal adenocarcinoma. However, the mechanistic link with established drivers of this disease remains in part elusive. In this study, using a new genetically engineered mouse model overexpressing constitutively active mouse form of GLI2 and a combination of genome-wide assays, we provide evidence of a novel mechanism underlying the interplay between KRAS, a major driver of pancreatic ductal adenocarcinoma development, and GLI2 to control oncogenic gene expression. These mice, also expressing KrasG12D, show significantly reduced median survival rate and accelerated tumorigenesis compared with the KrasG12D only expressing mice. Analysis of the mechanism using RNA sequencing demonstrate higher levels of GLI2 targets, particularly tumor growth-promoting genes, including Ccnd1, N-Myc, and Bcl2, in KrasG12D mutant cells. Furthermore, chromatin immunoprecipitation sequencing studies showed that in these cells KrasG12D increases the levels of trimethylation of lysine 4 of the histone 3 (H3K4me3) at the promoter of GLI2 targets without affecting significantly the levels of other major active chromatin marks. Importantly, Gli2 knockdown reduces H3K4me3 enrichment and gene expression induced by mutant Kras. In summary, we demonstrate that Gli2 plays a significant role in pancreatic carcinogenesis by acting as a downstream effector of KrasG12D to control gene expression.
Collapse
Affiliation(s)
- Ashley N. Sigafoos
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Ezequiel J. Tolosa
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Ryan M. Carr
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Maite G. Fernandez-Barrena
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Luciana L. Almada
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - David R. Pease
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Tara L. Hogenson
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Glancis L. Raja Arul
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Fatemeh Mousavi
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada.
- Department of Oncology, University of Western Ontario, London, Canada.
| | - Sandhya Sen
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Renzo E. Vera
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - David L. Marks
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Luis F. Flores
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Kayla C. LaRue-Nolan
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Chen Wu
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - William R. Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | - Anne M. Vrabel
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Hugues Sicotte
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | - Erin L. Schenk
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Thomas C. Smyrk
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota.
| | - Lizhi Zhang
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota.
| | - Kari G. Rabe
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | - Ann L. Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota.
| | | | - Eric Chevet
- Université de Rennes, CEDEX, Rennes, France.
| | | | | | - Marina P. di Magliano
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.
| | - Sherine F. Elsawa
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire.
| | - Christopher L. Pin
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada.
- Department of Oncology, University of Western Ontario, London, Canada.
| | - Junhao Mao
- University of Massachusetts Medical School, Worcester, Massachusetts.
| | | | - Martin E. Fernandez-Zapico
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
2
|
Coppola A, Grasso D, Fontana F, Piacentino F, Minici R, Laganà D, Ierardi AM, Carrafiello G, D’Angelo F, Carcano G, Venturini M. Innovative Experimental Ultrasound and US-Related Techniques Using the Murine Model in Pancreatic Ductal Adenocarcinoma: A Systematic Review. J Clin Med 2023; 12:7677. [PMID: 38137745 PMCID: PMC10743777 DOI: 10.3390/jcm12247677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/24/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a cancer with one of the highest mortality rates in the world. Several studies have been conductedusing preclinical experiments in mice to find new therapeutic strategies. Experimental ultrasound, in expert hands, is a safe, multifaceted, and relatively not-expensive device that helps researchers in several ways. In this systematic review, we propose a summary of the applications of ultrasonography in a preclinical mouse model of PDAC. Eighty-eight studies met our inclusion criteria. The included studies could be divided into seven main topics: ultrasound in pancreatic cancer diagnosis and progression (n: 21); dynamic contrast-enhanced ultrasound (DCE-US) (n: 5); microbubble ultra-sound-mediated drug delivery; focused ultrasound (n: 23); sonodynamic therapy (SDT) (n: 7); harmonic motion elastography (HME) and shear wave elastography (SWE) (n: 6); ultrasound-guided procedures (n: 9). In six cases, the articles fit into two or more sections. In conclusion, ultrasound can be a really useful, eclectic, and ductile tool in different diagnostic areas, not only regarding diagnosis but also in therapy, pharmacological and interventional treatment, and follow-up. All these multiple possibilities of use certainly represent a good starting point for the effective and wide use of murine ultrasonography in the study and comprehensive evaluation of pancreatic cancer.
Collapse
Affiliation(s)
- Andrea Coppola
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Dario Grasso
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Federico Fontana
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Filippo Piacentino
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Roberto Minici
- Radiology Unit, Dulbecco University Hospital, 88100 Catanzaro, Italy; (R.M.)
| | - Domenico Laganà
- Radiology Unit, Dulbecco University Hospital, 88100 Catanzaro, Italy; (R.M.)
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Anna Maria Ierardi
- Radiology Unit, IRCCS Ca Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Fabio D’Angelo
- Department of Medicine and Surgery, Insubria University, 21100 Varese, Italy;
- Orthopedic Surgery Unit, ASST Sette Laghi, 21100 Varese, Italy
| | - Giulio Carcano
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
- Emergency and Transplant Surgery Department, ASST Sette Laghi, 21100 Varese, Italy
| | - Massimo Venturini
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy (M.V.)
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| |
Collapse
|
3
|
GLI1 interaction with p300 modulates SDF1 expression in cancer-associated fibroblasts to promote pancreatic cancer cells migration. Biochem J 2023; 480:225-241. [PMID: 36734208 DOI: 10.1042/bcj20220521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/04/2023]
Abstract
Carcinoma-associated fibroblasts (CAFs) play an important role in the progression of multiple malignancies. Secretion of cytokines and growth factors underlies the pro-tumoral effect of CAFs. Although this paracrine function has been extensively documented, the molecular mechanisms controlling the expression of these factors remain elusive. In this study, we provide evidence of a novel CAF transcriptional axis regulating the expression of SDF1, a major driver of cancer cell migration, involving the transcription factor GLI1 and histone acetyltransferase p300. We demonstrate that conditioned media from CAFs overexpressing GLI1 induce the migration of pancreatic cancer cells, and this effect is impaired by an SDF1-neutralizing antibody. Using a combination of co-immunoprecipitation, proximity ligation assay and chromatin immunoprecipitation assay, we further demonstrate that GLI1 and p300 physically interact in CAFs to co-occupy and drive SDF1 promoter activity. Mapping experiments highlight the requirement of GLI1 N-terminal for the interaction with p300. Importantly, knockdowns of both GLI1 and p300 reduce SDF1 expression. Further analysis shows that knockdown of GLI1 decreases SDF1 promoter activity, p300 recruitment, and levels of its associated histone marks (H4ac, H3K27ac, and H3K14ac). Finally, we show that the integrity of two GLI binding sites in the SDF1 promoter is required for p300 recruitment. Our findings define a new role for the p300-GLI1 complex in the regulation of SDF1, providing new mechanistic insight into the molecular events controlling pancreatic cancer cells migration.
Collapse
|
4
|
Aberrant transcription factors in the cancers of the pancreas. Semin Cancer Biol 2022; 86:28-45. [PMID: 36058426 DOI: 10.1016/j.semcancer.2022.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/15/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022]
Abstract
Transcription factors (TFs) are essential for proper activation of gene set during the process of organogenesis, differentiation, lineage specificity. Reactivation or dysregulation of TFs regulatory networks could lead to deformation of organs, diseases including various malignancies. Currently, understanding the mechanism of oncogenesis became necessity for the development of targeted therapeutic strategy for different cancer types. It is evident that many TFs go awry in cancers of the pancreas such as pancreatic ductal adenocarcinoma (PDAC) and pancreatic neuroendocrine neoplasms (PanNENs). These mutated or dysregulated TFs abnormally controls various signaling pathways in PDAC and PanNENs including RTK, PI3K-PTEN-AKT-mTOR, JNK, TGF-β/SMAD, WNT/β-catenin, SHH, NOTCH and VEGF which in turn regulate different hallmarks of cancer. Aberrant regulation of such pathways have been linked to the initiation, progression, metastasis, and resistance in pancreatic cancer. As of today, a number of TFs has been identified as crucial regulators of pancreatic cancer and a handful of them shown to have potential as therapeutic targets in pre-clinical and clinical settings. In this review, we have summarized the current knowledge on the role and therapeutic usefulness of TFs in PDAC and PanNENs.
Collapse
|
5
|
Zhuang L, Yao Y, Peng L, Cui F, Chen C, Zhang Y, Sun L, Yu Q, Lin K. Silencing GS Homeobox 2 Alleviates Gemcitabine Resistance in Pancreatic Cancer Cells by Activating SHH/GLI1 Signaling Pathway. Dig Dis Sci 2022; 67:3773-3782. [PMID: 34623580 DOI: 10.1007/s10620-021-07262-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
Sonic hedgehog (SHH) signaling pathway and glioma-associated oncogene homolog 1 (GLI1) play important roles in the initiation and progression of pancreatic ductal adenocarcinoma (PDAC). GS homeobox 2 (GSX2, formerly GSH2) is a downstream target of SHH signaling, but its role in pancreatic cancer remains unclear. This study evaluates the role of GSH2 in the development and drug resistance of pancreatic cancer. Both cell culture and xenograft mouse model were used. Immunohistochemistry, Western blotting and quantitative RT-PCR were used to examine the expression of GSH2 and other related molecules. CCK8 assay was used to test the cell proliferation, and flow cytometry used to examine cell apoptosis upon gemcitabine treatment. It was found that GSH2 is overexpressed in human pancreatic cancer tissues and cells. The expression of SHH and GLI1 was reversely correlated with GSH2 in pancreatic cancer cells. SHH and GLI1 have protein-protein interactions with GSH2. GSH2 silencing in pancreatic cancer cells inhibited cell proliferation, migration and invasion, increased cell apoptosis and sensitized pancreatic cancer cells to gemcitabine treatment. Furthermore, in vivo study demonstrated that silencing GSH2 increased the efficacy of gemcitabine-based treatment. Our results indicate that GSH2 is overexpressed in pancreatic cancer. GSH2 silencing in pancreatic cancer alleviates gemcitabine resistance by activating SHH/GLI1 pathway. Thus, targeting GSH2 in PDAC could be a novel cancer therapeutic strategy.
Collapse
Affiliation(s)
- Lu Zhuang
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
- Shanghai Hongkou District Jiaxing Road Subdistrict Community Healthcare Service Center, 1 Hongguan Road, Shanghai, 200086, China
| | - Yao Yao
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200030, China
| | - Lisi Peng
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Fang Cui
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Cui Chen
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yang Zhang
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Liqi Sun
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Qihong Yu
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Kun Lin
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| |
Collapse
|
6
|
Scales MK, Velez-Delgado A, Steele NG, Schrader HE, Stabnick AM, Yan W, Mercado Soto NM, Nwosu ZC, Johnson C, Zhang Y, Salas-Escabillas DJ, Menjivar RE, Maurer HC, Crawford HC, Bednar F, Olive KP, Pasca di Magliano M, Allen BL. Combinatorial Gli activity directs immune infiltration and tumor growth in pancreatic cancer. PLoS Genet 2022; 18:e1010315. [PMID: 35867772 PMCID: PMC9348714 DOI: 10.1371/journal.pgen.1010315] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/03/2022] [Accepted: 06/27/2022] [Indexed: 01/16/2023] Open
Abstract
Proper Hedgehog (HH) signaling is essential for embryonic development, while aberrant HH signaling drives pediatric and adult cancers. HH signaling is frequently dysregulated in pancreatic cancer, yet its role remains controversial, with both tumor-promoting and tumor-restraining functions reported. Notably, the GLI family of HH transcription factors (GLI1, GLI2, GLI3), remain largely unexplored in pancreatic cancer. We therefore investigated the individual and combined contributions of GLI1-3 to pancreatic cancer progression. At pre-cancerous stages, fibroblast-specific Gli2/Gli3 deletion decreases immunosuppressive macrophage infiltration and promotes T cell infiltration. Strikingly, combined loss of Gli1/Gli2/Gli3 promotes macrophage infiltration, indicating that subtle changes in Gli expression differentially regulate immune infiltration. In invasive tumors, Gli2/Gli3 KO fibroblasts exclude immunosuppressive myeloid cells and suppress tumor growth by recruiting natural killer cells. Finally, we demonstrate that fibroblasts directly regulate macrophage and T cell migration through the expression of Gli-dependent cytokines. Thus, the coordinated activity of GLI1-3 directs the fibroinflammatory response throughout pancreatic cancer progression.
Collapse
Affiliation(s)
- Michael K. Scales
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ashley Velez-Delgado
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Nina G. Steele
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Hannah E. Schrader
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Anna M. Stabnick
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Wei Yan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Nayanna M. Mercado Soto
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Zeribe C. Nwosu
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Rosa E. Menjivar
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - H. Carlo Maurer
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York city, New York, United States of America
- Internal Medicine II, School of Medicine, Technische Universität München, Munich, Germany
| | - Howard C. Crawford
- Department of Surgery, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Filip Bednar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kenneth P. Olive
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York city, New York, United States of America
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York city, New York, United States of America
| | - Marina Pasca di Magliano
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Benjamin L. Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
7
|
Koeniger A, Brichkina A, Nee I, Dempwolff L, Hupfer A, Galperin I, Finkernagel F, Nist A, Stiewe T, Adhikary T, Diederich W, Lauth M. Activation of Cilia-Independent Hedgehog/GLI1 Signaling as a Novel Concept for Neuroblastoma Therapy. Cancers (Basel) 2021; 13:cancers13081908. [PMID: 33921042 PMCID: PMC8071409 DOI: 10.3390/cancers13081908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Elevated GLI1 expression levels are associated with improved survival in NB patients and GLI1 overexpression exerts tumor-suppressive traits in cultured NB cells. However, NB cells are protected from increased GLI1 levels as they have lost the ability to form primary cilia and transduce Hedgehog signals. This study identifies an isoxazole (ISX) molecule with primary cilia-independent GLI1-activating properties, which blocks NB cell growth. Mechanistically, ISX combines the removal of GLI3 repressor and the inhibition of class I HDACs, providing proof-of-principle evidence that small molecule-mediated activation of GLI1 could be harnessed therapeutically in the future. Abstract Although being rare in absolute numbers, neuroblastoma (NB) represents the most frequent solid tumor in infants and young children. Therapy options and prognosis are comparably good for NB patients except for the high risk stage 4 class. Particularly in adolescent patients with certain genetic alterations, 5-year survival rates can drop below 30%, necessitating the development of novel therapy approaches. The developmentally important Hedgehog (Hh) pathway is involved in neural crest differentiation, the cell type being causal in the etiology of NB. However, and in contrast to its function in some other cancer types, Hedgehog signaling and its transcription factor GLI1 exert tumor-suppressive functions in NB, rendering GLI1 an interesting new candidate for anti-NB therapy. Unfortunately, the therapeutic concept of pharmacological Hh/GLI1 pathway activation is difficult to implement as NB cells have lost primary cilia, essential organelles for Hh perception and activation. In order to bypass this bottleneck, we have identified a GLI1-activating small molecule which stimulates endogenous GLI1 production without the need for upstream Hh pathway elements such as Smoothened or primary cilia. This isoxazole compound potently abrogates NB cell proliferation and might serve as a starting point for the development of a novel class of NB-suppressive molecules.
Collapse
Affiliation(s)
- Anke Koeniger
- Center for Tumor- and Immune Biology, Department of Gastroenterology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (A.B.); (A.H.); (I.G.)
| | - Anna Brichkina
- Center for Tumor- and Immune Biology, Department of Gastroenterology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (A.B.); (A.H.); (I.G.)
| | - Iris Nee
- Department of Medicinal Chemistry and Center for Tumor- and Immune Biology, Philipps University Marburg, 35043 Marburg, Germany; (I.N.); (L.D.); (W.D.)
| | - Lukas Dempwolff
- Department of Medicinal Chemistry and Center for Tumor- and Immune Biology, Philipps University Marburg, 35043 Marburg, Germany; (I.N.); (L.D.); (W.D.)
| | - Anna Hupfer
- Center for Tumor- and Immune Biology, Department of Gastroenterology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (A.B.); (A.H.); (I.G.)
| | - Ilya Galperin
- Center for Tumor- and Immune Biology, Department of Gastroenterology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (A.B.); (A.H.); (I.G.)
| | - Florian Finkernagel
- Center for Tumor- and Immune Biology, Bioinformatics Core Facility, Philipps University Marburg, 35043 Marburg, Germany;
| | - Andrea Nist
- Member of the German Center for Lung Research (DZL), Center for Tumor- and Immune Biology, Genomics Core Facility, Institute of Molecular Oncology, Philipps University Marburg, 35043 Marburg, Germany; (A.N.); (T.S.)
| | - Thorsten Stiewe
- Member of the German Center for Lung Research (DZL), Center for Tumor- and Immune Biology, Genomics Core Facility, Institute of Molecular Oncology, Philipps University Marburg, 35043 Marburg, Germany; (A.N.); (T.S.)
| | - Till Adhikary
- Institute for Biomedical Informatics and Biostatistics, Philipps University Marburg, 35043 Marburg, Germany;
| | - Wibke Diederich
- Department of Medicinal Chemistry and Center for Tumor- and Immune Biology, Philipps University Marburg, 35043 Marburg, Germany; (I.N.); (L.D.); (W.D.)
- Core Facility Medicinal Chemistry, Philipps University Marburg, 35043 Marburg, Germany
| | - Matthias Lauth
- Center for Tumor- and Immune Biology, Department of Gastroenterology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (A.B.); (A.H.); (I.G.)
- Correspondence:
| |
Collapse
|
8
|
Steele NG, Biffi G, Kemp SB, Zhang Y, Drouillard D, Syu L, Hao Y, Oni TE, Brosnan E, Elyada E, Doshi A, Hansma C, Espinoza C, Abbas A, The S, Irizarry-Negron V, Halbrook CJ, Franks NE, Hoffman MT, Brown K, Carpenter ES, Nwosu ZC, Johnson C, Lima F, Anderson MA, Park Y, Crawford HC, Lyssiotis CA, Frankel TL, Rao A, Bednar F, Dlugosz AA, Preall JB, Tuveson DA, Allen BL, Pasca di Magliano M. Inhibition of Hedgehog Signaling Alters Fibroblast Composition in Pancreatic Cancer. Clin Cancer Res 2021; 27:2023-2037. [PMID: 33495315 PMCID: PMC8026631 DOI: 10.1158/1078-0432.ccr-20-3715] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/17/2020] [Accepted: 01/14/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease characterized by an extensive fibroinflammatory stroma, which includes abundant cancer-associated fibroblast (CAF) populations. PDAC CAFs are heterogeneous, but the nature of this heterogeneity is incompletely understood. The Hedgehog pathway functions in PDAC in a paracrine manner, with ligands secreted by cancer cells signaling to stromal cells in the microenvironment. Previous reports investigating the role of Hedgehog signaling in PDAC have been contradictory, with Hedgehog signaling alternately proposed to promote or restrict tumor growth. In light of the newly discovered CAF heterogeneity, we investigated how Hedgehog pathway inhibition reprograms the PDAC microenvironment. EXPERIMENTAL DESIGN We used a combination of pharmacologic inhibition, gain- and loss-of-function genetic experiments, cytometry by time-of-flight, and single-cell RNA sequencing to study the roles of Hedgehog signaling in PDAC. RESULTS We found that Hedgehog signaling is uniquely activated in fibroblasts and differentially elevated in myofibroblastic CAFs (myCAF) compared with inflammatory CAFs (iCAF). Sonic Hedgehog overexpression promotes tumor growth, while Hedgehog pathway inhibition with the smoothened antagonist, LDE225, impairs tumor growth. Furthermore, Hedgehog pathway inhibition reduces myCAF numbers and increases iCAF numbers, which correlates with a decrease in cytotoxic T cells and an expansion in regulatory T cells, consistent with increased immunosuppression. CONCLUSIONS Hedgehog pathway inhibition alters fibroblast composition and immune infiltration in the pancreatic cancer microenvironment.
Collapse
Affiliation(s)
- Nina G Steele
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Giulia Biffi
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Samantha B Kemp
- Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, Michigan
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | | | - LiJyun Syu
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, New York
| | - Tobiloba E Oni
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Erin Brosnan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Ela Elyada
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Abhishek Doshi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Christa Hansma
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Carlos Espinoza
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Ahmed Abbas
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Stephanie The
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | | | - Christopher J Halbrook
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Nicole E Franks
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Megan T Hoffman
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Kristee Brown
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Eileen S Carpenter
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Zeribe C Nwosu
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Fatima Lima
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Michelle A Anderson
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Howard C Crawford
- Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Costas A Lyssiotis
- Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | | | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Michigan Institute of Data Science (MIDAS), University of Michigan, Ann Arbor, Michigan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Filip Bednar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | | | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Marina Pasca di Magliano
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
9
|
Oda SK, Anderson KG, Ravikumar P, Bonson P, Garcia NM, Jenkins CM, Zhuang S, Daman AW, Chiu EY, Bates BM, Greenberg PD. A Fas-4-1BB fusion protein converts a death to a pro-survival signal and enhances T cell therapy. J Exp Med 2020; 217:e20191166. [PMID: 32860705 PMCID: PMC7953733 DOI: 10.1084/jem.20191166] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 06/02/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022] Open
Abstract
Adoptive T cell therapy (ACT) with genetically modified T cells has shown impressive results against some hematologic cancers, but efficacy in solid tumors can be limited by restrictive tumor microenvironments (TMEs). For example, Fas ligand is commonly overexpressed in TMEs and induces apoptosis in tumor-infiltrating, Fas receptor-positive lymphocytes. We engineered immunomodulatory fusion proteins (IFPs) to enhance ACT efficacy, combining an inhibitory receptor ectodomain with a costimulatory endodomain to convert negative into positive signals. We developed a Fas-4-1BB IFP that replaces the Fas intracellular tail with costimulatory 4-1BB. Fas-4-1BB IFP-engineered murine T cells exhibited increased pro-survival signaling, proliferation, antitumor function, and altered metabolism in vitro. In vivo, Fas-4-1BB ACT eradicated leukemia and significantly improved survival in the aggressive KPC pancreatic cancer model. Fas-4-1BB IFP expression also enhanced primary human T cell function in vitro. Thus, Fas-4-1BB IFP expression is a novel strategy to improve multiple T cell functions and enhance ACT against solid tumors and hematologic malignancies.
Collapse
Affiliation(s)
- Shannon K. Oda
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Pranali Ravikumar
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Patrick Bonson
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Nicolas M. Garcia
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Cody M. Jenkins
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Summer Zhuang
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Andrew W. Daman
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Edison Y. Chiu
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Breanna M. Bates
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Philip D. Greenberg
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine/Oncology, University of Washington, Seattle, WA
| |
Collapse
|
10
|
Carr RM, Duma N, McCleary-Wheeler AL, Almada LL, Marks DL, Graham RP, Smyrk TC, Lowe V, Borad MJ, Kim G, Johnson GB, Allred JB, Yin J, Lim VS, Bekaii-Saab T, Ma WW, Erlichman C, Adjei AA, Fernandez-Zapico ME. Targeting of the Hedgehog/GLI and mTOR pathways in advanced pancreatic cancer, a phase 1 trial of Vismodegib and Sirolimus combination. Pancreatology 2020; 20:1115-1122. [PMID: 32778368 DOI: 10.1016/j.pan.2020.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES Preclinical data indicated a functional and molecular interaction between Hedgehog (HH)/GLI and PI3K-AKT-mTOR pathways promoting pancreatic ductal adenocarcinoma (PDAC). A phase I study was conducted of Vismodegib and Sirolimus combination to evaluate maximum tolerated dose (MTD) and preliminary anti-tumor efficacy. METHODS Cohort I included advanced solid tumors patients following a traditional 3 + 3 design. Vismodegib was orally administered at 150 mg daily with Sirolimus starting at 3 mg daily, increasing to 6 mg daily at dose level 2. Cohort II included only metastatic PDAC patients. Anti-tumor efficacy was evaluated every two cycles and target assessment at pre-treatment and after a single cycle. RESULTS Nine patient were enrolled in cohort I and 22 patients in cohort II. Twenty-eight patients were evaluated for dose-limiting toxicities (DLTs). One DLT was observed in each cohort, consisting of grade 2 mucositis and grade 3 thrombocytopenia. The MTD for Vismodegib and Sirolimus were 150 mg daily and 6 mg daily, respectively. The most common grade 3-4 toxicities were fatigue, thrombocytopenia, dehydration, and infections. A total of 6 patients had stable disease. No partial or complete responses were observed. Paired biopsy analysis before and after the first cycle in cohort II consistently demonstrated reduced GLI1 expression. Conversely, GLI and mTOR downstream targets were not significantly affected. CONCLUSIONS The combination of Vismodegib and Sirolimus was well tolerated. Clinical benefit was limited to stable disease in a subgroup of patients. Targeting efficacy demonstrated consistent partial decreases in HH/GLI signaling with limited impact on mTOR signaling. These findings conflict with pre-clinical models and warrant further investigations.
Collapse
Affiliation(s)
- Ryan M Carr
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, USA; Department of Medical Oncology, Department of Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55902, USA
| | - Narjust Duma
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Angela L McCleary-Wheeler
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - David L Marks
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Rondell P Graham
- Department of Laboratory Medicine Pathology, Mayo Clinic, Rochester, MN, USA
| | - Thomas C Smyrk
- Department of Laboratory Medicine Pathology, Mayo Clinic, Rochester, MN, USA
| | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Mitesh J Borad
- Division of Hematology-Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - George Kim
- Division of Hematology-Oncology, The George Washington University, Washington, DC, USA
| | | | - Jacob B Allred
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jun Yin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Vun-Sin Lim
- Department of Medical Oncology, Department of Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55902, USA
| | - Tanios Bekaii-Saab
- Division of Hematology-Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Wen We Ma
- Department of Medical Oncology, Department of Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55902, USA
| | - Charles Erlichman
- Department of Medical Oncology, Department of Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55902, USA
| | - Alex A Adjei
- Department of Medical Oncology, Department of Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55902, USA.
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, USA; Department of Medical Oncology, Department of Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55902, USA.
| |
Collapse
|
11
|
Guo JC, Zhang P, Zhou L, You L, Liu QF, Zhang ZG, Sun B, Liang ZY, Lu J, Yuan D, Tan AD, Sun J, Liao Q, Dai MH, Xiao GG, Li S, Zhang TP. Prognostic and predictive value of a five-molecule panel in resected pancreatic ductal adenocarcinoma: A multicentre study. EBioMedicine 2020; 55:102767. [PMID: 32361251 PMCID: PMC7195527 DOI: 10.1016/j.ebiom.2020.102767] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/15/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) has a devastating prognosis. The performance of clinicopathologic parameters and molecules as prognostic factors remains limited and inconsistent. The present study aimed to construct a multi-molecule biomarker panel to more accurately predict post-resectional prognosis of PDAC patients. Methods Firstly, a novel computational strategy integrating prognostic evidence from omics and literature on the basis of bioinformatics prediction (CIPHER) to generate the network, was designed to systematically identify potential high-confidence PDAC-related prognostic candidates. After specimens from 605 resected PDAC patients were retrospectively collected, 23 candidates were detected immunohistochemically in tissue-microarrays for the development cohort to construct a multi-molecule panel. Lastly, the panel was validated in two independent cohorts. Findings According to the constructed five-molecule panel, disease-specific survival (DSS) was significantly poorer in high-risk patients than in low-risk ones in development cohort (HR 2.15, 95%CI 1.51–3.05, P<0.0001; AUC 0.67). In two validation cohorts, similar significant differences between the two groups were also observed (HR 3.18 and 3.31, 95%CI 1.89–5.37 and 1.78–6.16, All P<0.0001; AUC 0.72 and 0.73). In multivariate analyses, this panel was the sole prognosticator that was significant in each cohort. Furthermore, its predictive power for long-term survival, higher than its individual constituents, could be largely enhanced by combination with traditional clinicopathological variables. Finally, adjuvant chemotherapy (ACT) correlated with better DSS only in high-risk patients, uni- and multi-variately, in all the cohorts. Interpretation The novel prognostic panel developed by a systematically network-based strategy presents strong ability in prediction of post-resectional survival of PDAC patients. Furthermore, panel-defined high-risk patients might benefit more from ACT.
Collapse
Affiliation(s)
- Jun-Chao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.
| | - Peng Zhang
- MOE Key Laboratory of Bioinformatics, TCM-X Center/Bioinformatics Division, BNRIST/Department of Automation, Tsinghua University, Beijing, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Qiao-Fei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jun Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Da Yuan
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Ai-Di Tan
- MOE Key Laboratory of Bioinformatics, TCM-X Center/Bioinformatics Division, BNRIST/Department of Automation, Tsinghua University, Beijing, China
| | - Jian Sun
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Meng-Hua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Gary Guishan Xiao
- School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Shao Li
- MOE Key Laboratory of Bioinformatics, TCM-X Center/Bioinformatics Division, BNRIST/Department of Automation, Tsinghua University, Beijing, China.
| | - Tai-Ping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.
| |
Collapse
|
12
|
USP5 promotes tumorigenesis and progression of pancreatic cancer by stabilizing FoxM1 protein. Biochem Biophys Res Commun 2017; 492:48-54. [PMID: 28807830 DOI: 10.1016/j.bbrc.2017.08.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 08/10/2017] [Indexed: 02/08/2023]
Abstract
Increased ubiquitin-specific protease 5 (USP5) has been associated with tumorigenesis of malignancy including glioblastoma, melanoma and hepatocellular carcinoma. However, the role of USP5 in tumorigenesis of pancreatic ductal adenocarcinoma (PDAC) has not been studied yet. In this study, we demonstrated that USP5 was significantly upregulated in a panel of PDAC cell lines and correlated with FoxM1 protein expression. USP5 knockdown inhibited proliferation of PANC-1 and SW1990, two PDAC cell lines. In the mouse xenografted pancreatic tumor model, suppression of USP5 significantly decreased tumor growth, correlated with down regulation of FoxM1. Additionally, we found that overexpression of USP5 stabilized the FoxM1 protein in PDAC cells. Overexpression of USP5 extended the half-life of FoxM1. Knockdown of USP5 in PANC-1 cells decreased FoxM1 protein level while the proteasome inhibitor MG-132 treatment restored FoxM1 expression. We also found that endogenous USP5 was coimmunoprecipitated with an endogenous FoxM1 from PANC-1 cells while FoxM1 was also coimmunoprecipitated with USP5. Furthermore, we also confirmed that USP5 regulated proliferation of PDAC via FoxM1 by rescuing the inhibitory effect of USP5 knockdown with ectopic expression of FoxM1 in USP5-depleted cells. Taken together, our study demonstrates that USP5 plays a critical role in tumorigenesis and progression of pancreatic cancer by stabilizing FoxM1 protein, and provides a rationale for USP5 being a potential therapeutic approach against PDAC.
Collapse
|
13
|
Aberrant GLI1 Activation in DNA Damage Response, Carcinogenesis and Chemoresistance. Cancers (Basel) 2015; 7:2330-51. [PMID: 26633513 PMCID: PMC4695894 DOI: 10.3390/cancers7040894] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/12/2015] [Accepted: 11/20/2015] [Indexed: 12/18/2022] Open
Abstract
The canonical hedgehog (HH) pathway is a multicomponent signaling cascade (HH, protein patched homolog 1 (PTCH1), smoothened (SMO)) that plays a pivotal role during embryonic development through activation of downstream effector molecules, namely glioma-associated oncogene homolog 1 (GLI1), GLI2 and GLI3. Activation of GLIs must be tightly regulated as they modulate target genes which control tissue patterning, stem cell maintenance, and differentiation during development. However, dysregulation or mutations in HH signaling leads to genomic instability (GI) and various cancers, for example, germline mutation in PTCH1 lead to Gorlin syndrome, a condition where patients develop numerous basal cell carcinomas and rarely rhabdomyosarcoma (RMS). Activating mutations in SMO have also been recognized in sporadic cases of medulloblastoma and SMO is overexpressed in many other cancers. Recently, studies in several human cancers have shown that GLI1 expression is independent from HH ligand and canonical intracellular signaling through PTCH and SMO. In fact, this aberrantly regulated GLI1 has been linked to several non-canonical oncogenic growth signals such as Kirsten rat sarcoma viral oncogene homolog (KRAS), avian myelocytomatosis virus oncogene cellular homolog (C-MYC), transforming growth factor β (TGFβ), wingless-type MMTV integration site family (WNT) and β-catenin. Recent studies from our lab and other independent studies demonstrate that aberrantly expressed GLI1 influences the integrity of several DNA damage response and repair signals, and if altered, these networks can contribute to GI and impact tumor response to chemo- and radiation therapies. Furthermore, the ineffectiveness of SMO inhibitors in clinical studies argues for the development of GLI1-specific inhibitors in order to develop effective therapeutic modalities to treat these tumors. In this review, we focus on summarizing current understanding of the molecular, biochemical and cellular basis for aberrant GLI1 expression and discuss GLI1-mediated HH signaling on DNA damage responses, carcinogenesis and chemoresistance.
Collapse
|
14
|
Gopinathan A, Morton JP, Jodrell DI, Sansom OJ. GEMMs as preclinical models for testing pancreatic cancer therapies. Dis Model Mech 2015; 8:1185-200. [PMID: 26438692 PMCID: PMC4610236 DOI: 10.1242/dmm.021055] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is the most common form of pancreatic tumour, with a very limited survival rate and currently no available disease-modifying treatments. Despite recent advances in the production of genetically engineered mouse models (GEMMs), the development of new therapies for pancreatic cancer is still hampered by a lack of reliable and predictive preclinical animal models for this disease. Preclinical models are vitally important for assessing therapies in the first stages of the drug development pipeline, prior to their transition to the clinical arena. GEMMs carry mutations in genes that are associated with specific human diseases and they can thus accurately mimic the genetic, phenotypic and physiological aspects of human pathologies. Here, we discuss different GEMMs of human pancreatic cancer, with a focus on the Lox-Stop-Lox (LSL)-Kras(G12D); LSL-Trp53(R172H); Pdx1-cre (KPC) model, one of the most widely used preclinical models for this disease. We describe its application in preclinical research, highlighting its advantages and disadvantages, its potential for predicting clinical outcomes in humans and the factors that can affect such outcomes, and, finally, future developments that could advance the discovery of new therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Aarthi Gopinathan
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | | | - Duncan I Jodrell
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| |
Collapse
|
15
|
Wang H, Maitra A, Wang H. The emerging roles of F-box proteins in pancreatic tumorigenesis. Semin Cancer Biol 2015; 36:88-94. [PMID: 26384530 DOI: 10.1016/j.semcancer.2015.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 09/13/2015] [Indexed: 11/24/2022]
Abstract
The role of F-box proteins in pancreatic tumorigenesis is emerging owing to their pivotal and indispensable roles in cell differentiation, cell cycle regulation and proliferation. In this review, we will focus on β-TrCP (β-transducin repeat-containing protein) and two other prototypical mammalian F-box proteins, Fbxw7 and Fbxw8, in pancreatic tumorigenesis and progression. We will highlight the functions and regulation of these F-box proteins, their respective substrates and cross-talks with other key signaling pathways, such as the Ras-Raf-Mek-Erk, Hedgehog, NFκB, TGF-β, Myc and HPK1 signaling pathways in pancreatic cancer.
Collapse
Affiliation(s)
- Hua Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, United States
| | - Anirban Maitra
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, United States; Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, United States
| | - Huamin Wang
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, United States; Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, United States.
| |
Collapse
|
16
|
Involvement of JNK and caspase activation in hoiamide A-induced neurotoxicity in neocortical neurons. Mar Drugs 2015; 13:903-19. [PMID: 25675001 PMCID: PMC4344608 DOI: 10.3390/md13020903] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 01/24/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
The frequent occurrence of Moorea producens (formerly Lyngbya majuscula) blooms has been associated with adverse effects on human health. Hoiamide A is a structurally unique cyclic depsipeptide isolated from an assemblage of the marine cyanobacteria M. producens and Phormidiumgracile. We examined the influence of hoiamide A on neurite outgrowth in neocortical neurons and found that it suppressed neurite outgrowth with an IC50 value of 4.89 nM. Further study demonstrated that hoiamide A stimulated lactic acid dehydrogenase (LDH) efflux, nuclear condensation and caspase-3 activity with EC50 values of 3.66, 2.55 and 4.33 nM, respectively. These data indicated that hoiamide A triggered a unique neuronal death profile that involves both necrotic and apoptotic mechanisms. The similar potencies and similar time-response relationships between LDH efflux and caspase-3 activation/nuclear condensation suggested that both necrosis and apoptosis may derive from interaction with a common molecular target. The broad-spectrum caspase inhibitor, Z-VAD-FMK completely inhibited hoiamide A-induced neurotoxicity. Additionally, hoiamide A stimulated JNK phosphorylation, and a JNK inhibitor attenuated hoiamide A-induced neurotoxicity. Collectively, these data demonstrate that hoiamide A-induced neuronal death requires both JNK and caspase signaling pathways. The potent neurotoxicity and unique neuronal cell death profile of hoiamide A represents a novel neurotoxic chemotype from marine cyanobacteria.
Collapse
|
17
|
Hogenson TL, Lauth M, Pasca diMagliano M, Fernandez-Zapico ME. Back to the drawing board: Re-thinking the role of GLI1 in pancreatic carcinogenesis. F1000Res 2014; 3:238. [PMID: 25352983 PMCID: PMC4207242 DOI: 10.12688/f1000research.5324.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2016] [Indexed: 11/20/2022] Open
Abstract
Aberrant activation of the transcription factor GLI1, a central effector of the Hedgehog (HH) pathway, is associated with several malignancies, including pancreatic ductal adenocarcinoma (PDAC), one of most deadly human cancers. GLI1 has been described as an oncogene in PDAC, making it a promising target for drug therapy. Surprisingly, clinical trials targeting HH/GLI1 axis in advanced PDAC were unsuccessful, leaving investigators questioning the mechanism behind these failures. Recent evidence suggests the loss of GLI1 in the later stages of PDAC may actually accelerate disease. This indicates GLI1 may play a dual role in PDAC, acting as an oncogene in the early stages of disease and a tumor-suppressor in the late stages.
Collapse
Affiliation(s)
- Tara L. Hogenson
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthias Lauth
- Institute of Molecular Biology and Tumor Research, Philipps University, Marburg, 35043, Germany
| | | | | |
Collapse
|
18
|
Hogenson TL, Lauth M, Pasca diMagliano M, Fernandez-Zapico ME. Back to the drawing board: Re-thinking the role of GLI1 in pancreatic carcinogenesis. F1000Res 2014; 3:238. [PMID: 25352983 PMCID: PMC4207242 DOI: 10.12688/f1000research.5324.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/07/2014] [Indexed: 03/26/2024] Open
Abstract
Aberrant activation of the transcription factor GLI1, a central effector of the Hedgehog (HH) pathway, is associated with several malignancies, including pancreatic ductal adenocarcinoma (PDAC), one of most deadly human cancers. GLI1 has been described as an oncogene in PDAC, making it a promising target for drug therapy. Surprisingly, clinical trials targeting HH/GLI1 axis in advanced PDAC were unsuccessful, leaving investigators questioning the mechanism behind these failures. Recent evidence suggests the loss of GLI1 in the later stages of PDAC may actually accelerate disease. This indicates GLI1 may play a dual role in PDAC, acting as an oncogene in the early stages of disease and a tumor-suppressor in the late stages.
Collapse
Affiliation(s)
- Tara L. Hogenson
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthias Lauth
- Institute of Molecular Biology and Tumor Research, Philipps University, Marburg, 35043, Germany
| | | | | |
Collapse
|