1
|
Yang J, Lewis JS, Zi J, Andl T, Lee E, Andl CD, Liu Q, Beauchamp RD, Means AL. Interaction of the tumor suppressor SMAD4 and WNT signaling in progression to oral squamous cell carcinoma. J Pathol 2024; 264:4-16. [PMID: 38922866 PMCID: PMC11300146 DOI: 10.1002/path.6318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024]
Abstract
SMAD4 is a tumor suppressor mutated or silenced in multiple cancers, including oral cavity squamous cell carcinoma (OSCC). Human clinical samples and cell lines, mouse models and organoid culture were used to investigate the role that SMAD4 plays in progression from benign disease to invasive OSCC. Human OSCC lost detectable SMAD4 protein within tumor epithelium in 24% of cases, and this loss correlated with worse progression-free survival independent of other major clinical and pathological features. A mouse model engineered for KrasG12D expression in the adult oral epithelium induced benign papillomas, however the combination of KrasG12D with loss of epithelial Smad4 expression resulted in rapid development of invasive carcinoma with features of human OSCC. Examination of regulatory pathways in 3D organoid cultures of SMAD4+ and SMAD4- mouse tumors with Kras mutation found that either loss of SMAD4 or inhibition of TGFβ signaling upregulated the WNT pathway and altered the extracellular matrix. The gene signature of the mouse tumor organoids lacking SMAD4 was highly similar to the gene signature of human head and neck squamous cell carcinoma. In summary, this work has uncovered novel mechanisms by which SMAD4 acts as a tumor suppressor in OSCC. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jing Yang
- Dept. of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - James S. Lewis
- Dept. of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Jinghuan Zi
- Dept. of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL
| | - Ethan Lee
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Dept. of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Claudia D. Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL
| | - Qi Liu
- Dept. of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Robert D. Beauchamp
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Dept. of Surgery, Vanderbilt University Medical Center, Nashville, TN
- Dept. of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, TN
| | - Anna L. Means
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Dept. of Surgery, Vanderbilt University Medical Center, Nashville, TN
- Dept. of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
2
|
Golivi Y, Kumari S, Farran B, Alam A, Peela S, Nagaraju GP. Small molecular inhibitors: Therapeutic strategies for pancreatic cancer. Drug Discov Today 2024; 29:104053. [PMID: 38849028 DOI: 10.1016/j.drudis.2024.104053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Pancreatic cancer (PC), a disease with high heterogeneity and a dense stromal microenvironment, presents significant challenges and a bleak prognosis. Recent breakthroughs have illuminated the crucial interplay among RAS, epidermal growth factor receptor (EGFR), and hedgehog pathways in PC progression. Small molecular inhibitors have emerged as a potential solution with their advantages of oral administration and the ability to target intracellular and extracellular sites effectively. However, despite the US FDA approving over 100 small-molecule targeted antitumor drugs, challenges such as low response rates and drug resistance persist. This review delves into the possibility of using small molecules to treat persistent or spreading PC, highlighting the challenges and the urgent need for a diverse selection of inhibitors to develop more effective treatment strategies.
Collapse
Affiliation(s)
- Yuvasri Golivi
- Department of Bioscience and Biotechnology, Banasthali University, Banasthali, RJ 304 022, India
| | - Seema Kumari
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GIS, GITAM, Visakhapatnam, Andhra Pradesh 530045, India
| | - Batoul Farran
- Department of Hematology and Oncology, Henry Ford Health, Detroit, MI 48202, USA
| | - Afroz Alam
- Department of Bioscience and Biotechnology, Banasthali University, Banasthali, RJ 304 022, India
| | - Sujatha Peela
- Department of Biotechnology, Dr. B. R. Ambedkar University, Srikakulam, Andhra Pradesh, 532001, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
3
|
Hedegger K, Blutke A, Hommel T, Auer KE, Nataraj NB, Lindzen M, Yarden Y, Dahlhoff M. Trapping all ERBB ligands decreases pancreatic lesions in a murine model of pancreatic ductal adenocarcinoma. Mol Oncol 2023; 17:2415-2431. [PMID: 37341059 PMCID: PMC10620123 DOI: 10.1002/1878-0261.13473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/11/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest of cancers. Attempts to develop targeted therapies still need to be established. Some oncogenic mechanisms in PDAC carcinogenesis harness the EGFR/ERBB receptor family. To explore the effects on pancreatic lesions, we attempted simultaneous blockade of all ERBB ligands in a PDAC mouse model. To this end, we engineered a molecular decoy, TRAP-FC , comprising the ligand-binding domains of both EGFR and ERBB4 and able to trap all ERBB ligands. Next, we generated a transgenic mouse model (CBATRAP/0 ) expressing TRAP-FC ubiquitously under the control of the chicken-beta-actin promoter and crossed these mice with KRASG12D/+ mice (Kras) to generate Trap/Kras mice. The resulting mice displayed decreased emergence of spontaneous pancreatic lesion areas and exhibited reduced RAS activity and decreased activities of ERBBs, with the exception of ERBB4, which showed increased activity. To identify the involved receptor(s), we employed CRISPR/Cas9 DNA editing to singly delete each ERBB receptor in the human pancreatic carcinoma cell line Panc-1. Ablation of each ERBB family member, especially the loss of EGFR or ERBB2/HER2, altered signaling downstream of the other three ERBB receptors and decreased cell proliferation, migration, and tumor growth. We conclude that simultaneously blocking the entire ERBB receptor family is therapeutically more effective than individually inhibiting only one receptor or ligand in terms of reducing pancreatic tumor burden. In summary, trapping all ERBB ligands can reduce pancreatic lesion area and RAS activity in a murine model of pancreatic adenocarcinoma; hence, it might represent a promising approach to treat PDAC in patients.
Collapse
Affiliation(s)
- Kathrin Hedegger
- Institute of Molecular Animal Breeding and Biotechnology, Gene CenterLMU MünchenGermany
| | - Andreas Blutke
- Institute of Veterinary Pathology, Center for Clinical Veterinary MedicineLMU MünchenGermany
| | - Theresa Hommel
- Institute of in vivo and in vitro ModelsUniversity of Veterinary MedicineViennaAustria
| | - Kerstin E. Auer
- Institute of in vivo and in vitro ModelsUniversity of Veterinary MedicineViennaAustria
| | - Nishanth B. Nataraj
- Department of Immunology and Regenerative BiologyWeizmann Institute of ScienceRehovotIsrael
- Bugworks Research Inc, CCAMPBengaluruIndia
| | - Moshit Lindzen
- Department of Immunology and Regenerative BiologyWeizmann Institute of ScienceRehovotIsrael
| | - Yosef Yarden
- Department of Immunology and Regenerative BiologyWeizmann Institute of ScienceRehovotIsrael
| | - Maik Dahlhoff
- Institute of in vivo and in vitro ModelsUniversity of Veterinary MedicineViennaAustria
| |
Collapse
|
4
|
Liou GY, Fleming Martinez AK, Döppler HR, Bastea LI, Storz P. Inflammatory and alternatively activated macrophages independently induce metaplasia but cooperatively drive pancreatic precancerous lesion growth. iScience 2023; 26:106820. [PMID: 37250781 PMCID: PMC10212997 DOI: 10.1016/j.isci.2023.106820] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/16/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
The innate immune system has a key role in pancreatic cancer initiation, but the specific contribution of different macrophage populations is still ill-defined. While inflammatory (M1) macrophages have been shown to drive acinar-to-ductal metaplasia (ADM), a cancer initiating event, alternatively activated (M2) macrophages have been attributed to lesion growth and fibrosis. Here, we determined cytokines and chemokines secreted by both macrophage subtypes. Then, we analyzed their role in ADM initiation and lesion growth, finding that while M1 secrete TNF, CCL5, and IL-6 to drive ADM, M2 induce this dedifferentiation process via CCL2, but the effects are not additive. This is because CCL2 induces ADM by generating ROS and upregulating EGFR signaling, thus using the same mechanism as cytokines from inflammatory macrophages. Therefore, while effects on ADM are not additive between macrophage polarization types, both act synergistically on the growth of low-grade lesions by activating different MAPK pathways.
Collapse
Affiliation(s)
- Geou-Yarh Liou
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Biological Sciences, Center for Cancer Research & Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | | | - Heike R. Döppler
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ligia I. Bastea
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
5
|
Yang K, Yang T, Yu J, Li F, Zhao X. Integrated transcriptional analysis reveals macrophage heterogeneity and macrophage-tumor cell interactions in the progression of pancreatic ductal adenocarcinoma. BMC Cancer 2023; 23:199. [PMID: 36864399 PMCID: PMC9983236 DOI: 10.1186/s12885-023-10675-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease harboring significant microenvironment heterogeneity, especially for the macrophages. Tumor-associated macrophages (TAMs) orchestrate PDAC malignancy, but their dynamics during disease progression remains poorly understood. There is a pressing need to identify the molecular mechanism underlying tumor-macrophage interactions and thus design novel therapeutic strategies. METHODS Herein, we developed an insilico computational method incorporating bulk and single-cell transcriptome profiling to characterize macrophage heterogeneity. CellPhoneDB algorithm was applied to infer macrophage-tumor interaction networks, whereas pseudotime trajectory for dissecting cell evolution and dynamics. RESULTS We demonstrated myeloid compartment was an interactive hub of tumor microenvironment (TME) essential for PDAC progression. Dimensionality reduction classified seven clusters within the myeloid cells wherein five subsets of macrophages were characterized by diverse cell states and functionality. Remarkably, tissue-resident macrophages and inflammatory monocyte were identified as potential sources of TAMs. Further, we uncovered several ligand-receptor pairs lining tumor cells and macrophages. Among them, HBEGF-CD44, HBEGF-EGFR, LGALS9-CD44, LGALS9-MET, and GRN-EGFR were correlated with worse overall survival. Notably, as in vitro experiments indicated, TAM-derived HBEGF promoted proliferation and invasion of the pancreatic cancer cell line. CONCLUSION Together, our work deciphered a comprehensive single-cell atlas of the macrophage compartment of PDAC and provided novel macrophage-tumor interaction features with potential value in developing targeted immunotherapies and molecular diagnostics for predicting patient outcome.
Collapse
Affiliation(s)
- Kaidi Yang
- Department of Oncology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, 57200, Hainan Province, P.R. China. .,Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P. R. China.
| | - Tongxin Yang
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army General Hospital, Sanya, 57200 Hainan Province P.R. China
| | - Jian Yu
- grid.73113.370000 0004 0369 1660Department of Health Statistics, Naval Medical University, Shanghai, 200433 PR China
| | - Fang Li
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army General Hospital, Sanya, 57200 Hainan Province P.R. China
| | - Xiang Zhao
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, P. R. China.
| |
Collapse
|
6
|
Ye L, Shi S, Chen W. Innate immunity in pancreatic cancer: Lineage tracing and function. Front Immunol 2023; 13:1081919. [PMID: 36726981 PMCID: PMC9884680 DOI: 10.3389/fimmu.2022.1081919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/28/2022] [Indexed: 01/17/2023] Open
Abstract
Increasingly, patients with gastrointestinal tumors can benefit from immunotherapy, but not patients with pancreatic cancer. While this lack of benefit has been attributed to lower T-cell infiltration in pancreatic cancer, other studies have demonstrated the presence of numerous T cells in pancreatic cancer, suggesting another mechanism for the poor efficacy of immunotherapy. Single-cell RNA sequencing studies on the pancreatic cancer immune microenvironment have demonstrated the predominance of innate immune cells (e.g., macrophages, dendritic cells, mast cells, and innate immune lymphoid cells). Therefore, in-depth research on the source and function of innate immune lymphocytes in pancreatic cancer could guide pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Longyun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Saimeng Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wei Chen
- Institute of Clinical Medicine Research, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
7
|
Bhatia R, Bhyravbhatla N, Kisling A, Li X, Batra SK, Kumar S. Cytokines chattering in pancreatic ductal adenocarcinoma tumor microenvironment. Semin Cancer Biol 2022; 86:499-510. [PMID: 35346801 PMCID: PMC9510605 DOI: 10.1016/j.semcancer.2022.03.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME) consists of multiple cell types interspersed by dense fibrous stroma. These cells communicate through low molecular weight signaling molecules called cytokines. The cytokines, through their receptors, facilitate PDAC initiation, progression, metastasis, and distant colonization of malignant cells. These signaling mediators secreted from tumor-associated macrophages, and cancer-associated fibroblasts in conjunction with oncogenic Kras mutation initiate acinar to ductal metaplasia (ADM), resulting in the appearance of early preneoplastic lesions. Further, M1- and M2-polarized macrophages provide proinflammatory conditions and promote deposition of extracellular matrix, whereas myofibroblasts and T-lymphocytes, such as Th17 and T-regulatory cells, create a fibroinflammatory and immunosuppressive environment with a significantly reduced cytotoxic T-cell population. During PDAC progression, cytokines regulate the expression of various oncogenic regulators such as NFκB, c-myc, growth factor receptors, and mucins resulting in the formation of high-grade PanIN lesions, epithelial to mesenchymal transition, invasion, and extravasation of malignant cells, and metastasis. During metastasis, PDAC cells colonize at the premetastatic niche created in the liver, and lung, an organotropic function primarily executed by cytokines in circulation or loaded in the exosomes from the primary tumor cells. The indispensable contribution of these cytokines at every stage of PDAC tumorigenesis makes them exciting candidates in combination with immune-, chemo- and targeted radiation therapy.
Collapse
Affiliation(s)
- Rakesh Bhatia
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Namita Bhyravbhatla
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew Kisling
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiaoqi Li
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Omaha, NE, USA.
| |
Collapse
|
8
|
Afzal J, Du W, Novin A, Liu Y, Wali K, Murthy A, Garen A, Wagner G. Paracrine HB-EGF signaling reduce enhanced contractile and energetic state of activated decidual fibroblasts by rebalancing SRF-MRTF-TCF transcriptional axis. Front Cell Dev Biol 2022; 10:927631. [PMID: 36147738 PMCID: PMC9485834 DOI: 10.3389/fcell.2022.927631] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022] Open
Abstract
Multiple parallels exist between placentation and cancer dissemination at molecular, cellular, and anatomical levels, presenting placentation as a unique model to mechanistically understand the onset of cancer metastasis. In humans, interaction of placenta and the endometrium results eventually in deep invasion of placental extravillous trophoblasts (EVTs) into the maternal stroma, a process similar to stromal trespass by disseminating carcinoma cells. In anticipation of implantation, endometrial fibroblasts (ESFs) undergo a process called decidualization during the secretory phase of the menstrual cycle. Decidualization, among other substantial changes associated with ESF differentiation, also involves a component of fibroblast activation, and myofibroblast transformation. Here, using traction force microscopy, we show that increased cellular contractility in decidualized ESFs is reversed after interaction with EVTs. We also report here the large changes in energetic state of ESFs upon decidualization, showing increased oxidative phosphorylation, mitochondrial competency and ATP generation, as well as enhanced aerobic glycolysis, presenting mechanical contractility and energetic state as new functional hallmarks for decidualization. These energetic changes accompanying the marked increase in contractile force generation in decidualization were reduced in the presence of EVTs. We also show that increase in decidual contractility and mechanical resistance to invasion is achieved by SRF-MRTF transcriptional activation, achieved via increased phosphorylation of fibroblast-specific myosin light chain 9 (MYL9). EVT induced paracrine secretion of Heparin Binding Epidermal Growth Factor (HBEGF), a potent MAPK activator, which shifts the balance of SRF association away from MRTF based transcription, reducing decidual ESF contractility and mechanical resistance to placental invasion. Our results identify a new axis of intercellular communication in the placental bed modulating stromal force generation and resistance to invasion with concurrent downregulation of cellular energetics. These findings have important implications for implantation related disorders, as well as stromal control of cancer dissemination.
Collapse
Affiliation(s)
- Junaid Afzal
- Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- *Correspondence: Junaid Afzal, ; Kshitiz,
| | - Wenqiang Du
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, United States
| | - Ashkan Novin
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, United States
| | - Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, United States
| | - Khadija Wali
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, United States
| | - Anarghya Murthy
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, United States
| | - Ashley Garen
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, United States
| | - Gunter Wagner
- Department of Ecology and Evolution, Yale University West Campus, West Haven, CT, United States
- Systems Biology Institute, Yale University, West Haven, CT, United States
| |
Collapse
|
9
|
Vidimar V, Park M, Stubbs CK, Ingram NK, Qiang W, Zhang S, Gursel D, Melnyk RA, Satchell KJF. Proteolytic pan-RAS Cleavage Leads to Tumor Regression in Patient-derived Pancreatic Cancer Xenografts. Mol Cancer Ther 2022; 21:810-820. [PMID: 35247912 PMCID: PMC9933180 DOI: 10.1158/1535-7163.mct-21-0550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/12/2021] [Accepted: 02/22/2022] [Indexed: 11/16/2022]
Abstract
The lack of effective RAS inhibition represents a major unmet medical need in the treatment of pancreatic ductal adenocarcinoma (PDAC). Here, we investigate the anticancer activity of RRSP-DTB, an engineered biologic that cleaves the Switch I of all RAS isoforms, in KRAS-mutant PDAC cell lines and patient-derived xenografts (PDX). We first demonstrate that RRSP-DTB effectively engages RAS and impacts downstream ERK signaling in multiple KRAS-mutant PDAC cell lines inhibiting cell proliferation at picomolar concentrations. We next tested RRSP-DTB in immunodeficient mice bearing KRAS-mutant PDAC PDXs. Treatment with RRSP-DTB led to ≥95% tumor regression after 29 days. Residual tumors exhibited disrupted tissue architecture, increased fibrosis and fewer proliferating cells compared with controls. Intratumoral levels of phospho-ERK were also significantly lower, indicating in vivo target engagement. Importantly, tumors that started to regrow without RRSP-DTB shrank when treatment resumed, demonstrating resistance to RRSP-DTB had not developed. Tracking persistence of the toxin activity following intraperitoneal injection showed that RRSP-DTB is active in sera from immunocompetent mice for at least 1 hour, but absent after 16 hours, justifying use of daily dosing. Overall, we report that RRSP-DTB strongly regresses hard-to-treat KRAS-mutant PDX models of pancreatic cancer, warranting further development of this pan-RAS biologic for the management of RAS-addicted tumors.
Collapse
Affiliation(s)
- Vania Vidimar
- Department of Microbiology and Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Minyoung Park
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Caleb K Stubbs
- Department of Microbiology and Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Nana K Ingram
- Department of Microbiology and Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Wenan Qiang
- Center for Developmental Therapeutics, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
- Department of Obstetrics and Gynecology (Reproductive Science in Medicine), Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Pathology Core Facility, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Research Center, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Shanshan Zhang
- Pathology Core Facility, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Demirkan Gursel
- Pathology Core Facility, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Roman A Melnyk
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Karla J F Satchell
- Department of Microbiology and Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Robert H. Lurie Comprehensive Cancer Research Center, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
10
|
Poh AR, Ernst M. Tumor-Associated Macrophages in Pancreatic Ductal Adenocarcinoma: Therapeutic Opportunities and Clinical Challenges. Cancers (Basel) 2021; 13:cancers13122860. [PMID: 34201127 PMCID: PMC8226457 DOI: 10.3390/cancers13122860] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Macrophages are a major component of the pancreatic tumor microenvironment, and their increased abundance is associated with poor patient survival. Given the multi-faceted role of macrophages in promoting pancreatic tumor development and progression, these cells represent promising targets for anti-cancer therapy. Abstract Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignant disease with a 5-year survival rate of less than 10%. Macrophages are one of the earliest infiltrating cells in the pancreatic tumor microenvironment, and are associated with an increased risk of disease progression, recurrence, metastasis, and shorter overall survival. Pre-clinical studies have demonstrated an unequivocal role of macrophages in PDAC by contributing to chronic inflammation, cancer cell stemness, desmoplasia, immune suppression, angiogenesis, invasion, metastasis, and drug resistance. Several macrophage-targeting therapies have also been investigated in pre-clinical models, and include macrophage depletion, inhibiting macrophage recruitment, and macrophage reprogramming. However, the effectiveness of these drugs in pre-clinical models has not always translated into clinical trials. In this review, we discuss the molecular mechanisms that underpin macrophage heterogeneity within the pancreatic tumor microenvironment, and examine the contribution of macrophages at various stages of PDAC progression. We also provide a comprehensive update of macrophage-targeting therapies that are currently undergoing clinical evaluation, and discuss clinical challenges associated with these treatment modalities in human PDAC patients.
Collapse
|
11
|
Marincola Smith P, Choksi YA, Markham NO, Hanna DN, Zi J, Weaver CJ, Hamaamen JA, Lewis KB, Yang J, Liu Q, Kaji I, Means AL, Beauchamp RD. Colon epithelial cell TGFβ signaling modulates the expression of tight junction proteins and barrier function in mice. Am J Physiol Gastrointest Liver Physiol 2021; 320:G936-G957. [PMID: 33759564 PMCID: PMC8285585 DOI: 10.1152/ajpgi.00053.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 01/31/2023]
Abstract
Defective barrier function is a predisposing factor in inflammatory bowel disease (IBD) and colitis-associated cancer (CAC). Although TGFβ signaling defects have been associated with IBD and CAC, few studies have examined the relationship between TGFβ and intestinal barrier function. Here, we examine the role of TGFβ signaling via SMAD4 in modulation of colon barrier function. The Smad4 gene was conditionally deleted in the intestines of adult mice and intestinal permeability assessed using an in vivo 4 kDa FITC-Dextran (FD4) permeability assay. Mouse colon was isolated for gene expression (RNA-sequencing), Western blot, and immunofluorescence analysis. In vitro colon organoid culture was utilized to assess junction-related gene expression by qPCR and transepithelial resistance (TER). In silico analyses of human IBD and colon cancer databases were performed. Mice lacking intestinal expression of Smad4 demonstrate increased colonic permeability to FD4 without gross mucosal damage. mRNA/protein expression analyses demonstrate significant increases in Cldn2/Claudin 2 and Cldn8/Claudin 8, and decreases in Cldn3, Cldn4, and Cldn7/Claudin 7 with intestinal SMAD4 loss in vivo without changes in Claudin protein localization. TGFβ1/BMP2 treatment of polarized SMAD4+ colonoids increases TER. Cldn2, Cldn4, Cldn7, and Cldn8 are regulated by canonical TGFβ signaling, and TGFβ-dependent regulation of these genes is dependent on nascent RNA transcription (Cldn2, Cldn4, Cldn8) but not nascent protein translation (Cldn4, Cldn8). Human IBD/colon cancer specimens demonstrate decreased SMAD4, CLDN4, CLDN7, and CLDN8 and increased CLDN2 compared with healthy controls. Canonical TGFβ signaling modulates the expression of tight junction proteins and barrier function in mouse colon.NEW & NOTEWORTHY We demonstrate that canonical TGFβ family signaling modulates the expression of critical tight junction proteins in colon epithelial cells, and that expression of these tight junction proteins is associated with maintenance of colon epithelial barrier function in mice.
Collapse
Affiliation(s)
- Paula Marincola Smith
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Yash A Choksi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Nicholas O Markham
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David N Hanna
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jinghuan Zi
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Connie J Weaver
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jalal A Hamaamen
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Keeli B Lewis
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jing Yang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Izumi Kaji
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna L Means
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - R Daniel Beauchamp
- Section of Surgical Sciences, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
12
|
Hedegger K, Algül H, Lesina M, Blutke A, Schmid RM, Schneider MR, Dahlhoff M. Unraveling ERBB network dynamics upon betacellulin signaling in pancreatic ductal adenocarcinoma in mice. Mol Oncol 2020; 14:1653-1669. [PMID: 32335999 PMCID: PMC7400790 DOI: 10.1002/1878-0261.12699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/17/2020] [Accepted: 04/06/2020] [Indexed: 12/28/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) will soon belong to the top three cancer killers. The only approved specific PDAC therapy targets the epidermal growth factor receptor (EGFR). Although EGFR is a crucial player in PDAC development, EGFR-based therapy is disappointing. In this study, we evaluated the role of the EGFR ligand betacellulin (BTC) in PDAC. The expression of BTC was investigated in human pancreatic cancer specimen. Then, we generated a BTC knockout mouse model by CRISPR/Cas9 technology and a BTC overexpression model. Both models were crossed with the Ptf1aCre/+ ;KRASG12D/+ (KC) mouse model (B-/- KC or BKC, respectively). In addition, EGFR, ERBB2, and ERBB4 were investigated by the pancreas-specific deletion of each receptor using the Cre-loxP system. Tumor initiation and progression were analyzed in all mouse lines, and the underlying molecular biology of PDAC was investigated at different time points. BTC is expressed in human and murine PDAC. B-/- KC mice showed a decelerated PDAC progression, associated with decreased EGFR activation. BKC mice developed severe PDAC with a poor survival rate. The dramatically increased BTC-mediated tumor burden was EGFR-dependent, but also ERBB4 and ERBB2 were involved in PDAC development or progression, as depletion of EGFR, ERBB2, or ERBB4 significantly improved the survival rate of BTC-mediated PDAC. BTC increases PDAC tumor burden dramatically by enhanced RAS activation. EGFR signaling, ERBB2 signaling, and ERBB4 signaling are involved in accelerated PDAC development mediated by BTC indicating that targeting the whole ERBB family, instead of a single receptor, is a promising strategy for the development of future PDAC therapies.
Collapse
Affiliation(s)
- Kathrin Hedegger
- Institute of Molecular Animal Breeding and BiotechnologyGene Center of the LMU MunichGermany
| | - Hana Algül
- Second Department of Internal MedicineKlinikum rechts der IsarTechnical University of MunichGermany
| | - Marina Lesina
- Second Department of Internal MedicineKlinikum rechts der IsarTechnical University of MunichGermany
| | - Andreas Blutke
- Research Unit Analytical PathologyHelmholtz Zentrum MünchenNeuherbergGermany
| | - Roland M. Schmid
- Second Department of Internal MedicineKlinikum rechts der IsarTechnical University of MunichGermany
| | - Marlon R. Schneider
- Institute of Molecular Animal Breeding and BiotechnologyGene Center of the LMU MunichGermany
| | - Maik Dahlhoff
- Institute of Molecular Animal Breeding and BiotechnologyGene Center of the LMU MunichGermany
| |
Collapse
|
13
|
Hegde S, Krisnawan VE, Herzog BH, Zuo C, Breden MA, Knolhoff BL, Hogg GD, Tang JP, Baer JM, Mpoy C, Lee KB, Alexander KA, Rogers BE, Murphy KM, Hawkins WG, Fields RC, DeSelm CJ, Schwarz JK, DeNardo DG. Dendritic Cell Paucity Leads to Dysfunctional Immune Surveillance in Pancreatic Cancer. Cancer Cell 2020; 37:289-307.e9. [PMID: 32183949 PMCID: PMC7181337 DOI: 10.1016/j.ccell.2020.02.008] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/04/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022]
Abstract
Here, we utilized spontaneous models of pancreatic and lung cancer to examine how neoantigenicity shapes tumor immunity and progression. As expected, neoantigen expression during lung adenocarcinoma development leads to T cell-mediated immunity and disease restraint. By contrast, neoantigen expression in pancreatic ductal adenocarcinoma (PDAC) results in exacerbation of a fibro-inflammatory microenvironment that drives disease progression and metastasis. Pathogenic TH17 responses are responsible for this neoantigen-induced tumor progression in PDAC. Underlying these divergent T cell responses in pancreas and lung cancer are differences in infiltrating conventional dendritic cells (cDCs). Overcoming cDC deficiency in early-stage PDAC leads to disease restraint, while restoration of cDC function in advanced PDAC restores tumor-restraining immunity and enhances responsiveness to radiation therapy.
Collapse
Affiliation(s)
- Samarth Hegde
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Varintra E Krisnawan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett H Herzog
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chong Zuo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcus A Breden
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham D Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jack P Tang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John M Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kyung Bae Lee
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine A Alexander
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Buck E Rogers
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William G Hawkins
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Ryan C Fields
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Carl J DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Julie K Schwarz
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - David G DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA.
| |
Collapse
|
14
|
Yao Y, Gu Y, Yang M, Cao D, Wu F. The Gene Expression Biomarkers for Chronic Obstructive Pulmonary Disease and Interstitial Lung Disease. Front Genet 2019; 10:1154. [PMID: 31824564 PMCID: PMC6879656 DOI: 10.3389/fgene.2019.01154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023] Open
Abstract
COPD (chronic obstructive pulmonary disease) and ILD (interstitial lung disease) are two common respiratory diseases. They share similar clinical traits but require different therapeutic treatments. Identifying the biomarkers that are differentially expressed between them will not only help the diagnosis of COPD and ILD, but also provide candidate drug targets that may facilitate the development of new treatment for COPD and ILD. Due to the irreversible complex pathological changes of COPD, there are very limited therapeutic options for COPD patients. In this study, we analyzed the gene expression profiles of two datasets: one training dataset that includes 144 COPD patients and 194 ILD patients, and one test dataset that includes 75 COPD patients and 61 ILD patients. Advanced feature selection methods, mRMR (minimal Redundancy Maximal Relevance) and incremental feature selection (IFS), were applied to identify the 38-gene biomarker. An SVM (support vector machine) classifier was built based on the 38-gene biomarker. Its accuracy, sensitivity, and specificity on training dataset evaluated by leave one out cross-validation were 0.905, 0.896, and 0.912, respectively. And on independent test dataset, the accuracy, sensitivity, and specificity on were as great as and were 0.904, 0.933, and 0.869, respectively. The biological function analysis of the 38 genes indicated that many of them can be potential treatment targets that may benefit COPD and ILD patients.
Collapse
Affiliation(s)
- Yangwei Yao
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Yangyang Gu
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Meng Yang
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Dakui Cao
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Fengjie Wu
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| |
Collapse
|
15
|
Abstract
A common biomarker of pancreatic disease has a functional role in pathogenesis
Collapse
Affiliation(s)
- Christopher J Halbrook
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Howard C Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- The Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
16
|
Wen HJ, Gao S, Wang Y, Ray M, Magnuson MA, Wright CV, Di Magliano MP, Frankel TL, Crawford HC. Myeloid Cell-Derived HB-EGF Drives Tissue Recovery After Pancreatitis. Cell Mol Gastroenterol Hepatol 2019; 8:173-192. [PMID: 31125624 PMCID: PMC6661420 DOI: 10.1016/j.jcmgh.2019.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Pancreatitis is a major cause of morbidity and mortality and is a risk factor for pancreatic tumorigenesis. Upon tissue damage, an inflammatory response, made up largely of macrophages, provides multiple growth factors that promote repair. Here, we examine the molecular pathways initiated by macrophages to promote pancreas recovery from pancreatitis. METHODS To induce organ damage, mice were subjected to cerulein-induced experimental pancreatitis and analyzed at various times of recovery. CD11b-DTR mice were used to deplete myeloid cells. Hbegff/f;LysM-Cre mice were used to ablate myeloid cell-derived heparin-binding epidermal growth factor (EGF)-like growth factor (HB-EGF). To ablate EGFR specifically during recovery, pancreatitis was induced in Egfrf/f;Ptf1aFlpO/+;FSF-Rosa26CAG-CreERT2 mice followed by tamoxifen treatment. RESULTS Macrophages infiltrating the pancreas in experimental pancreatitis make high levels of HB-EGF. Both depletion of myeloid cells and ablation of myeloid cell HB-EGF delayed recovery from experimental pancreatitis, resulting from a decrease in cell proliferation and an increase in apoptosis. Mechanistically, ablation of myeloid cell HB-EGF impaired epithelial cell DNA repair, ultimately leading to cell death. Soluble HB-EGF induced EGFR nuclear translocation and methylation of histone H4, facilitating resolution of DNA damage in pancreatic acinar cells in vitro. Consistent with its role as the primary receptor of HB-EGF, in vivo ablation of EGFR from pancreatic epithelium during recovery from pancreatitis resulted in accumulation of DNA damage. CONCLUSIONS By using novel conditional knockout mouse models, we determined that HB-EGF derived exclusively from myeloid cells induces epithelial cell proliferation and EGFR-dependent DNA repair, facilitating pancreas healing after injury.
Collapse
Affiliation(s)
- Hui-Ju Wen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Shan Gao
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, China
| | - Yin Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Michael Ray
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Mark A. Magnuson
- Department of Molecular Physiology and Biophysics, Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee
| | | | - Marina Pasca Di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan,Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | | | - Howard C. Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan,Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan,Correspondence Address correspondence to: Howard Crawford, PhD, University of Michigan, 4304 Rogel Cancer Center, 1500 East Medical Center Drive, SPC 5936, Ann Arbor, Michigan 48109-5936. fax: (734) 647–9654.
| |
Collapse
|
17
|
Means AL, Freeman TJ, Zhu J, Woodbury LG, Marincola-Smith P, Wu C, Meyer AR, Weaver CJ, Padmanabhan C, An H, Zi J, Wessinger BC, Chaturvedi R, Brown TD, Deane NG, Coffey RJ, Wilson KT, Smith JJ, Sawyers CL, Goldenring JR, Novitskiy SV, Washington MK, Shi C, Beauchamp RD. Epithelial Smad4 Deletion Up-Regulates Inflammation and Promotes Inflammation-Associated Cancer. Cell Mol Gastroenterol Hepatol 2018; 6:257-276. [PMID: 30109253 PMCID: PMC6083016 DOI: 10.1016/j.jcmgh.2018.05.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/18/2018] [Indexed: 02/08/2023]
Abstract
Background & Aims Chronic inflammation is a predisposing condition for colorectal cancer. Many studies to date have focused on proinflammatory signaling pathways in the colon. Understanding the mechanisms that suppress inflammation, particularly in epithelial cells, is critical for developing therapeutic interventions. Here, we explored the roles of transforming growth factor β (TGFβ) family signaling through SMAD4 in colonic epithelial cells. Methods The Smad4 gene was deleted specifically in adult murine intestinal epithelium. Colitis was induced by 3 rounds of dextran sodium sulfate in drinking water, after which mice were observed for up to 3 months. Nontransformed mouse colonocyte cell lines and colonoid cultures and human colorectal cancer cell lines were analyzed for responses to TGFβ1 and bone morphogenetic protein 2. Results Dextran sodium sulfate treatment was sufficient to drive carcinogenesis in mice lacking colonic Smad4 expression, with resulting tumors bearing striking resemblance to human colitis-associated carcinoma. Loss of SMAD4 protein was observed in 48% of human colitis-associated carcinoma samples as compared with 19% of sporadic colorectal carcinomas. Loss of Smad4 increased the expression of inflammatory mediators within nontransformed mouse colon epithelial cells in vivo. In vitro analysis of mouse and human colonic epithelial cell lines and organoids indicated that much of this regulation was cell autonomous. Furthermore, TGFβ signaling inhibited the epithelial inflammatory response to proinflammatory cytokines. Conclusions TGFβ suppresses the expression of proinflammatory genes in the colon epithelium, and loss of its downstream mediator, SMAD4, is sufficient to initiate inflammation-driven colon cancer. Transcript profiling: GSE100082.
Collapse
Key Words
- AOM, azoxymethane
- APC, adenomatous polyposis coli
- BMP, bone morphogenetic protein
- CAC, colitis-associated carcinoma
- CCL20, Chemokine (C-C motif) ligand 20
- CRC, colorectal cancer
- CRISPR/Cas9, Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9
- Colitis-Associated Carcinoma
- DMEM, Dulbecco's modified Eagle medium
- DSS, dextran sodium sulfate
- FBS, fetal bovine serum
- FDR, false discovery rate
- GFP, green fluorescent protein
- HBSS, Hank's balanced salt solution
- IBD, inflammatory bowel disease
- IL, interleukin
- IMCS4fl/fl, immortalized mouse colonoctye cell line with loxP-flanked Smad4 alleles
- IMCS4null, immortalized mouse colonocyte cell line with deletion of the Smad4 alleles
- LPS, lipopolysaccharide
- PBS, phosphate-buffered saline
- PE, phycoerythrin
- R-SMAD, Receptor-SMAD
- SFG, retroviral vector
- STAT3, signal transducer and activator of transcription 3
- TGFβ
- TGFβ, transforming growth factor β
- TNF, tumor necrosis factor
- Tumor Necrosis Factor
- UC, ulcerative colitis
- WNT, wingless-type mouse mammary tumor virus integration site
- YAMC, young adult mouse colon epithelial cells
- mRNA, messenger RNA
- sgRNA, single-guide RNA
Collapse
Affiliation(s)
- Anna L. Means
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tanner J. Freeman
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jing Zhu
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Luke G. Woodbury
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Chao Wu
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anne R. Meyer
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Connie J. Weaver
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Hanbing An
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jinghuan Zi
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bronson C. Wessinger
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rupesh Chaturvedi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tasia D. Brown
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Natasha G. Deane
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J. Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Keith T. Wilson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - J. Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles L. Sawyers
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James R. Goldenring
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Sergey V. Novitskiy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - M. Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Chanjuan Shi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - R. Daniel Beauchamp
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
18
|
Chen CC, Chen LL, Li CP, Hsu YT, Jiang SS, Fan CS, Chua KV, Huang SX, Shyr YM, Chen LT, Huang TS. Myeloid-derived macrophages and secreted HSP90α induce pancreatic ductal adenocarcinoma development. Oncoimmunology 2018; 7:e1424612. [PMID: 29721383 PMCID: PMC5927518 DOI: 10.1080/2162402x.2018.1424612] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 12/31/2017] [Accepted: 01/03/2018] [Indexed: 12/23/2022] Open
Abstract
We detected a significant elevation of serum HSP90α levels in pancreatitis patients and even more in pancreatic ductal adenocarcinoma (PDAC) patients. However, there was no significant difference in the serum HSP90α levels between patients with early-stage and late-stage PDAC. To study whether elevation of serum HSP90α levels occurred early during PDAC development, we used LSL-KrasG12D/Pdx1-Cre transgenic mice as a studying model. Elevated serum HSP90α levels were detected before PDAC formation and an extracellular HSP90α (eHSP90α) inhibitor effectively prevented PDAC development. Both serum HSP90α level and pancreatic lesion were suppressed when the mice were administered a CD11b-antagonizing antibody, suggesting that CD11b+-myeloid cells were associated with eHSP90α levels and pancreatic carcinogenesis. Consistently, in CD11b-DTR-EGFP transgenic mouse model with CD11b+-myeloid cells depletion, serum HSP90α levels were suppressed and Panc-02 cell grafts failed to develop tumors. Macrophages and granulocytes are two common tissue-infiltrating CD11b+-myeloid cells. Duplex in situ hybridization assays suggested that macrophages were predominant HSP90α-expressing CD11b+-myeloid cells during PDAC development. Immunohistochemical and immunohistofluorescent staining results revealed that HSP90α-expressing cells included not only macrophages but also pancreatic ductal epithelial (PDE) cells. Cell culture studies also indicated that eHSP90α could be produced by macrophages and macrophage-stimulated PDE cells. Macrophages not only secreted significant amount of HSP90α, but also secreted interleukin-6 and interleukin-8 to induce a JAK2−STAT3 signaling axis in PDE cells, stimulating them to express and secrete HSP90α. eHSP90α further promoted cellular epithelial-mesenchymal transition, migration, and invasion in PDE cells. Besides myeloid cells, eHSP90α can be potentially taken as a target to suppress PDAC pathogenesis.
Collapse
Affiliation(s)
- Chia-Chi Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Li Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chung-Pin Li
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, and School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Hsu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chi-Shuan Fan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Kee Voon Chua
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Sheng-Xiang Huang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yi-Ming Shyr
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, and School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Tze-Sing Huang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.,Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
19
|
Zhang Y, Yan W, Mathew E, Kane KT, Brannon A, Adoumie M, Vinta A, Crawford HC, Pasca di Magliano M. Epithelial-Myeloid cell crosstalk regulates acinar cell plasticity and pancreatic remodeling in mice. eLife 2017; 6:27388. [PMID: 28980940 PMCID: PMC5690281 DOI: 10.7554/elife.27388] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 10/04/2017] [Indexed: 12/14/2022] Open
Abstract
Dedifferentiation of acini to duct-like cells occurs during the physiologic damage response in the pancreas, but this process can be co-opted by oncogenic Kras to drive carcinogenesis. Myeloid cells infiltrate the pancreas during the onset of pancreatic cancer, and promote carcinogenesis. Here, we show that the function of infiltrating myeloid cells is regulated by oncogenic Kras expressed in epithelial cells. In the presence of oncogenic Kras, myeloid cells promote acinar dedifferentiation and carcinogenesis. Upon inactivation of oncogenic Kras, myeloid cells promote re-differentiation of acinar cells, remodeling of the fibrotic stroma and tissue repair. Intriguingly, both aspects of myeloid cell activity depend, at least in part, on activation of EGFR/MAPK signaling, with different subsets of ligands and receptors in different target cells promoting carcinogenesis or repair, respectively. Thus, the cross-talk between epithelial cells and infiltrating myeloid cells determines the balance between tissue repair and carcinogenesis in the pancreas.
Collapse
Affiliation(s)
- Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, United States
| | - Wei Yan
- Department of Surgery, University of Michigan, Ann Arbor, United States.,Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Esha Mathew
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, United States
| | - Kevin T Kane
- Department of Surgery, University of Michigan, Ann Arbor, United States
| | - Arthur Brannon
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, United States.,Medical Scientist Training Program, University of Michigan, Ann Arbor, United States
| | - Maeva Adoumie
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, United States
| | - Alekya Vinta
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, United States
| | - Howard C Crawford
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, United States.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, United States
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, United States.,Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, United States.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, United States.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
20
|
Lai X, Wang M, McElyea SD, Sherman S, House M, Korc M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett 2017; 393:86-93. [PMID: 28232049 PMCID: PMC5386003 DOI: 10.1016/j.canlet.2017.02.019] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy that often presents clinically at an advanced stage and that may be confused with chronic pancreatitis (CP). Conversely, CP may be misdiagnosed as PDAC leading to unwarranted pancreas resection. Therefore, early PDAC diagnosis and clear differentiation between PDAC and CP are crucial for improved care. Exosomes are circulating microvesicles whose components can serve as cancer biomarkers. We compared exosomal glypican-1 (GPC1) and microRNA levels in normal control subjects and in patients with PDAC and CP. We report that exosomal GPC1 is not diagnostic for PDAC, whereas high exosomal levels of microRNA-10b, (miR-10b), miR-21, miR-30c, and miR-181a and low miR-let7a readily differentiate PDAC from normal control and CP samples. By contrast with GPC1, elevated exosomal miR levels decreased to normal values within 24 h following PDAC resection. All 29 PDAC cases exhibited significantly elevated exosomal miR-10b and miR-30c levels, whereas 8 cases had normal or slightly increased CA 19-9 levels. Thus, our exosomal miR signature is superior to exosomal GPC1 or plasma CA 19-9 levels in establishing a diagnosis of PDAC and differentiating between PDAC and CP.
Collapse
MESH Headings
- Area Under Curve
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- CA-19-9 Antigen/blood
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/surgery
- Case-Control Studies
- Chromatography, Liquid
- Diagnosis, Differential
- Exosomes/metabolism
- Gene Expression Profiling/methods
- Glypicans/blood
- Humans
- MicroRNAs/blood
- MicroRNAs/genetics
- Oligonucleotide Array Sequence Analysis
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/surgery
- Pancreatitis, Chronic/blood
- Pancreatitis, Chronic/diagnosis
- Pancreatitis, Chronic/genetics
- Predictive Value of Tests
- ROC Curve
- Reproducibility of Results
- Tandem Mass Spectrometry
- Transcriptome
Collapse
Affiliation(s)
- Xianyin Lai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mu Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Samantha Deitz McElyea
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stuart Sherman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Michael House
- Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA; Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Murray Korc
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
21
|
Wang J, Yao X, Huang J. New tricks for human farnesyltransferase inhibitor: cancer and beyond. MEDCHEMCOMM 2017; 8:841-854. [PMID: 30108801 PMCID: PMC6072492 DOI: 10.1039/c7md00030h] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 02/15/2017] [Indexed: 12/18/2022]
Abstract
Human protein farnesyltransferase (FTase) catalyzes the addition of a C15-farnesyl lipid group to the cysteine residue located in the COOH-terminal tetrapeptide motif of a variety of important substrate proteins, including well-known Ras protein superfamily. The farnesylation of Ras protein is required both for its normal physiological function, and for the transforming capacity of its oncogenic mutants. Over the last several decades, FTase inhibitors (FTIs) were developed to disrupt the farnesylation of oncogenic Ras as anti-cancer agents, and some of them have entered cancer clinical investigation. On the other hand, some substrates of FTase were demonstrated to be related with other human diseases, including Hutchinson-Gilford progeria syndrome, chronic hepatitis D, and cardiovascular diseases. In this review, we summarize the roles of FTase in malignant transformation, proliferation, apoptosis, angiogenesis, and metastasis of tumor cells, and the recently anticancer clinical research advances of FTIs. The therapeutic prospect of FTIs on several other human diseases is also discussed.
Collapse
Affiliation(s)
- Jingyuan Wang
- Shanghai Key Laboratory of New Drug Design , School of Pharmacy , East China University of Science and Technology , 130 Mei Long Road , Shanghai 200237 , China . ; Tel: (+86)21 64253681
| | - Xue Yao
- Shanghai Key Laboratory of New Drug Design , School of Pharmacy , East China University of Science and Technology , 130 Mei Long Road , Shanghai 200237 , China . ; Tel: (+86)21 64253681
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design , School of Pharmacy , East China University of Science and Technology , 130 Mei Long Road , Shanghai 200237 , China . ; Tel: (+86)21 64253681
| |
Collapse
|
22
|
Shin CH, Robinson JP, Sonnen JA, Welker AE, Yu DX, VanBrocklin MW, Holmen SL. HBEGF promotes gliomagenesis in the context of Ink4a/Arf and Pten loss. Oncogene 2017; 36:4610-4618. [PMID: 28368403 PMCID: PMC5552427 DOI: 10.1038/onc.2017.83] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 02/07/2017] [Accepted: 02/26/2017] [Indexed: 12/19/2022]
Abstract
Heparin-binding epidermal growth factor (EGF)-like growth factor (HBEGF) is a ligand for the epidermal growth factor receptor (EGFR), one of the most commonly amplified receptor tyrosine kinases (RTK) in glioblastoma. While HBEGF has been found to be expressed in a subset of malignant gliomas, its sufficiency for glioma initiation has not been evaluated. In this study, we demonstrate that HBEGF can initiate glioblastoma (GBM) in mice in the context of Ink4a/Arf and Pten loss, and that these tumors are similar to the classical GBM subtype observed in patients. Isogenic astrocytes from these mice showed activation not only of Egfr but also the RTK Axl in response to HBEGF stimulation. Deletion of either Egfr or Axl decreased the tumorigenic properties of HBEGF transformed cells; however only EGFR was able to rescue the phenotype in cells lacking both RTKs indicating that Egfr is required for activation of Axl in this context. Silencing of HBEGF in vivo resulted in tumor regression and significantly increased survival suggesting that HBEGF may be a clinically relevant target.
Collapse
Affiliation(s)
- C H Shin
- Huntsman Cancer Institute, University of Utah Health Sciences Center, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | - J P Robinson
- Hormel Institute, University of Minnesota, Austin, MN, USA
| | - J A Sonnen
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, UT, USA.,ARUP Laboratories, Salt Lake City, UT, USA
| | - A E Welker
- Huntsman Cancer Institute, University of Utah Health Sciences Center, University of Utah, Salt Lake City, UT, USA
| | - D X Yu
- Huntsman Cancer Institute, University of Utah Health Sciences Center, University of Utah, Salt Lake City, UT, USA.,Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | - M W VanBrocklin
- Huntsman Cancer Institute, University of Utah Health Sciences Center, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT, USA.,Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | - S L Holmen
- Huntsman Cancer Institute, University of Utah Health Sciences Center, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, UT, USA.,Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| |
Collapse
|
23
|
Fujiwara K, Yatabe M, Tofrizal A, Jindatip D, Yashiro T, Nagai R. Identification of M2 macrophages in anterior pituitary glands of normal rats and rats with estrogen-induced prolactinoma. Cell Tissue Res 2017; 368:371-378. [PMID: 28120110 DOI: 10.1007/s00441-016-2564-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/07/2016] [Indexed: 01/16/2023]
Abstract
Macrophages are present throughout the anterior pituitary gland. However, the features and function of macrophages in the gland are poorly understood. Recent studies have indicated that there are two main macrophage classes: M1 (classically activated) and M2 (alternatively activated). In this study, we examine whether both M1 and M2 macrophages are present in the anterior pituitary gland of rats. Our findings indicate that macrophages that are positive for CD68 (a pan-macrophage marker) were localized near capillaries in rat anterior pituitary gland. These macrophages were positive for iNOS or mannose receptor (MR), which are markers of M1 and M2 macrophages, respectively. To determine the morphological characteristics of M2 macrophages under pathological conditions, diethylstilbestrol (DES)-treated rats were used as an animal model of prolactinoma. After 2 weeks of DES treatment, a number of MR-immunopositive cells were present in the gland. Immunoelectron microscopy revealed that MR-immunopositive M2 macrophages had many small vesicles and moderately large vacuoles in cytoplasm. Phagosomes were sometimes present in cytoplasm. Interestingly, M2 macrophages in prolactinoma tissues did not usually exhibit distinct changes or differences during the normal, hyperplasia and adenoma stages. This study is the first to confirm that both M1 and M2 macrophages are present in the anterior pituitary gland of rats. Moreover, the number of M2 macrophages was greatly increased in rats with DES-induced prolactinoma. Future studies should attempt to characterize the functional role of M2 macrophages in the gland.
Collapse
Affiliation(s)
- Ken Fujiwara
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Megumi Yatabe
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Alimuddin Tofrizal
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Depicha Jindatip
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Tochigi, Japan.,Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Takashi Yashiro
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Tochigi, Japan.
| | | |
Collapse
|
24
|
Schneeweis C, Wirth M, Saur D, Reichert M, Schneider G. Oncogenic KRAS and the EGFR loop in pancreatic carcinogenesis-A connection to licensing nodes. Small GTPases 2017; 9:457-464. [PMID: 27880072 DOI: 10.1080/21541248.2016.1262935] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
EGFR signaling has a critical role in oncogenic KRAS-driven tumorigenesis of the pancreas, whereas it is dispensable in other organs. The complex signaling network engaged by oncogenic KRAS and its modulation by EGFR signaling, remains incompletely understood. In order to study early signaling events activated by oncogenic KRAS in the pancreas, we recently developed a novel model system based on murine primary pancreatic epithelial cells enabling the time-specific expression of mutant KrasG12D from its endogenous promoter. Here, we discuss our findings of a KrasG12D-induced autocrine EGFR loop, how this loop is integrated by the MYC oncogene, and point to possible translational implications.
Collapse
Affiliation(s)
- Christian Schneeweis
- a II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München , München , Germany
| | - Matthias Wirth
- a II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München , München , Germany
| | - Dieter Saur
- a II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München , München , Germany.,b German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) , Heidelberg , Germany
| | - Maximilian Reichert
- a II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München , München , Germany
| | - Günter Schneider
- a II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München , München , Germany.,b German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) , Heidelberg , Germany
| |
Collapse
|
25
|
Zhang Y, Velez-Delgado A, Mathew E, Li D, Mendez FM, Flannagan K, Rhim AD, Simeone DM, Beatty GL, Pasca di Magliano M. Myeloid cells are required for PD-1/PD-L1 checkpoint activation and the establishment of an immunosuppressive environment in pancreatic cancer. Gut 2017; 66:124-136. [PMID: 27402485 PMCID: PMC5256390 DOI: 10.1136/gutjnl-2016-312078] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/26/2016] [Accepted: 06/10/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pancreatic cancer is characterised by the accumulation of a fibro-inflammatory stroma. Within this stromal reaction, myeloid cells are a predominant population. Distinct myeloid subsets have been correlated with tumour promotion and unmasking of anti-tumour immunity. OBJECTIVE The goal of this study was to determine the effect of myeloid cell depletion on the onset and progression of pancreatic cancer and to understand the relationship between myeloid cells and T cell-mediated immunity within the pancreatic cancer microenvironment. METHODS Primary mouse pancreatic cancer cells were transplanted into CD11b-diphtheria toxin receptor (DTR) mice. Alternatively, the iKras* mouse model of pancreatic cancer was crossed into CD11b-DTR mice. CD11b+ cells (mostly myeloid cell population) were depleted by diphtheria toxin treatment during tumour initiation or in established tumours. RESULTS Depletion of myeloid cells prevented KrasG12D-driven pancreatic cancer initiation. In pre-established tumours, myeloid cell depletion arrested tumour growth and in some cases, induced tumour regressions that were dependent on CD8+ T cells. We found that myeloid cells inhibited CD8+ T-cell anti-tumour activity by inducing the expression of programmed cell death-ligand 1 (PD-L1) in tumour cells in an epidermal growth factor receptor (EGFR)/mitogen-activated protein kinases (MAPK)-dependent manner. CONCLUSION Our results show that myeloid cells support immune evasion in pancreatic cancer through EGFR/MAPK-dependent regulation of PD-L1 expression on tumour cells. Derailing this crosstalk between myeloid cells and tumour cells is sufficient to restore anti-tumour immunity mediated by CD8+ T cells, a finding with implications for the design of immune therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashley Velez-Delgado
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Esha Mathew
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Dongjun Li
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Flor M Mendez
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin Flannagan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew D Rhim
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Diane M Simeone
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gregory L Beatty
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
26
|
Abstract
Islet transplantation has set the ground for diabetes cell therapy and is still undergoing various developments that might improve clinical outcomes. Alternative sources for β-cell replacement strategies are now led by human pluripotent stem cells that demonstrate near-normal β-cell features after in vitro differentiation and which can reverse diabetes in mice. Yet, their propensity for tumor formation is still unresolved. The adult pancreas is suggested as a reservoir of facultative progenitors that could represent adequate candidates for β-cell engineering, either in vivo through pharmacological treatment or after expansion in culture. This review focuses on the latest developments in protocols aiming at de novo production of functional β cells.
Collapse
Affiliation(s)
- Philippe A Lysy
- Laboratoire de pédiatrie, institut de recherche expérimentale et clinique, université catholique de Louvain, 1000 Bruxelles, Belgique - Unité d'endocrinologie pédiatrique, cliniques universitaires Saint Luc, université catholique de Louvain, 1000 Bruxelles, Belgique
| |
Collapse
|
27
|
Abstract
Since insulin discovery, islet transplantation was the first protocol to show the possibility to cure patients with type 1 diabetes using low-risk procedures. The scarcity of pancreas donors triggered a burst of studies focused on the production of new β cells in vitro. These were rapidly dominated by pluripotent stem cells (PSCs) demonstrating diabetes-reversal potential in diabetic mice. Subsequent enthusiasm fostered a clinical trial with immunoisolated embryonic-derived pancreatic progenitors. Yet safety is the Achilles' heel of PSCs, and a whole branch of β cell engineering medicine focuses on transdifferentiation of adult pancreatic cells. New data showed the possibility to chemically stimulate acinar or α cells to undergo β cell neogenesis and provide opportunities to intervene in situ without the need for a transplant, at least after weighing benefits against systemic adverse effects. The current studies suggested the pancreas as a reservoir of facultative progenitors (e.g., in the duct lining) could be exploited ex vivo for expansion and β cell differentiation in timely fashion and without the hurdles of PSC use. Diabetes cell therapy is thus a growing field not only with great potential but also with many pitfalls to overcome for becoming fully envisioned as a competitor to the current treatment standards.
Collapse
Affiliation(s)
- Philippe A Lysy
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium.
- Pediatric Endocrinology Unit, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium.
| | - Elisa Corritore
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne M Sokal
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
28
|
Rosati A, Basile A, D'Auria R, d'Avenia M, De Marco M, Falco A, Festa M, Guerriero L, Iorio V, Parente R, Pascale M, Marzullo L, Franco R, Arra C, Barbieri A, Rea D, Menichini G, Hahne M, Bijlsma M, Barcaroli D, Sala G, di Mola FF, di Sebastiano P, Todoric J, Antonucci L, Corvest V, Jawhari A, Firpo MA, Tuveson DA, Capunzo M, Karin M, De Laurenzi V, Turco MC. BAG3 promotes pancreatic ductal adenocarcinoma growth by activating stromal macrophages. Nat Commun 2015; 6:8695. [PMID: 26522614 PMCID: PMC4659838 DOI: 10.1038/ncomms9695] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/21/2015] [Indexed: 12/16/2022] Open
Abstract
The incidence and death rate of pancreatic ductal adenocarcinoma (PDAC) have increased in recent years, therefore the identification of novel targets for treatment is extremely important. Interactions between cancer and stromal cells are critically involved in tumour formation and development of metastasis. Here we report that PDAC cells secrete BAG3, which binds and activates macrophages, inducing their activation and the secretion of PDAC supporting factors. We also identify IFITM-2 as a BAG3 receptor and show that it signals through PI3K and the p38 MAPK pathways. Finally, we show that the use of an anti-BAG3 antibody results in reduced tumour growth and prevents metastasis formation in three different mouse models. In conclusion, we identify a paracrine loop involved in PDAC growth and metastatic spreading, and show that an anti-BAG3 antibody has therapeutic potential.
Collapse
Affiliation(s)
- Alessandra Rosati
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Medicine and Surgery, University of Salerno,
Baronissi, Salerno
84081, Italy
| | - Anna Basile
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Medicine and Surgery, University of Salerno,
Baronissi, Salerno
84081, Italy
| | - Raffaella D'Auria
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Pharmacy, Division of Biomedicine “A.
Leone”, University of Salerno, Fisciano,
Salerno
84084, Italy
| | | | | | - Antonia Falco
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Pharmacy, Division of Biomedicine “A.
Leone”, University of Salerno, Fisciano,
Salerno
84084, Italy
| | - Michelina Festa
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Pharmacy, Division of Biomedicine “A.
Leone”, University of Salerno, Fisciano,
Salerno
84084, Italy
| | - Luana Guerriero
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Pharmacy, Division of Biomedicine “A.
Leone”, University of Salerno, Fisciano,
Salerno
84084, Italy
| | - Vittoria Iorio
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Medicine and Surgery, University of Salerno,
Baronissi, Salerno
84081, Italy
| | | | - Maria Pascale
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Pharmacy, Division of Biomedicine “A.
Leone”, University of Salerno, Fisciano,
Salerno
84084, Italy
| | - Liberato Marzullo
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Medicine and Surgery, University of Salerno,
Baronissi, Salerno
84081, Italy
| | - Renato Franco
- Pathology Unit, Istituto Nazionale Tumouri Fondazione
“G. Pascale”, Naples
81100, Italy
| | - Claudio Arra
- Animal facility, Istituto Nazionale Tumouri Fondazione
“G. Pascale”, Naples
81100, Italy
| | - Antonio Barbieri
- Animal facility, Istituto Nazionale Tumouri Fondazione
“G. Pascale”, Naples
81100, Italy
| | - Domenica Rea
- Animal facility, Istituto Nazionale Tumouri Fondazione
“G. Pascale”, Naples
81100, Italy
| | - Giulio Menichini
- Reconstructive Microsurgery, Department of Oncology, Careggi
University Hospital, Florence
50139, Italy
| | - Michael Hahne
- Institut de Génétique
Moléculaire de Montpellier, CNRS UMR5535,
Montpellier
34293, France
| | - Maarten Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Academic
Medical Center, University of Amsterdam, Amsterdam
1105AZ, The Netherlands
| | - Daniela Barcaroli
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche,
University “G. d'Annunzio” di Chieti-Pescara,
Centro Studi sull'Invecchiamento, CeSI-MeT, Chieti
66100, Italy
| | - Gianluca Sala
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche,
University “G. d'Annunzio” di Chieti-Pescara,
Centro Studi sull'Invecchiamento, CeSI-MeT, Chieti
66100, Italy
| | | | | | - Jelena Todoric
- Laboratory of Gene Regulation and Signal Transduction,
Departments of Pharmacology and Pathology, UCSD, School of Medicine,
San Diego, California
92093-0723, USA
| | - Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction,
Departments of Pharmacology and Pathology, UCSD, School of Medicine,
San Diego, California
92093-0723, USA
| | | | - Anass Jawhari
- CALIXAR, Bioparc, Bâtiment Laënnec,
Lyon
69008, France
| | - Matthew A Firpo
- Department of Surgery, Huntsman Cancer Institute, University of
Utah School of Medicine, Salt Lake City, Utah
84132, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring
Harbor, New York
11724, USA
| | - Mario Capunzo
- Department of Medicine and Surgery, University of Salerno,
Baronissi, Salerno
84081, Italy
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction,
Departments of Pharmacology and Pathology, UCSD, School of Medicine,
San Diego, California
92093-0723, USA
| | - Vincenzo De Laurenzi
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche,
University “G. d'Annunzio” di Chieti-Pescara,
Centro Studi sull'Invecchiamento, CeSI-MeT, Chieti
66100, Italy
| | - Maria Caterina Turco
- BIOUNIVERSA s.r.l., Fisciano, Salerno
84084, Italy
- Department of Medicine and Surgery, University of Salerno,
Baronissi, Salerno
84081, Italy
| |
Collapse
|
29
|
The Arkadia-ESRP2 axis suppresses tumor progression: analyses in clear-cell renal cell carcinoma. Oncogene 2015; 35:3514-23. [PMID: 26522722 PMCID: PMC5399154 DOI: 10.1038/onc.2015.412] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/17/2015] [Indexed: 01/01/2023]
Abstract
Tumor-specific alternative splicing is implicated in the progression of cancer, including clear-cell renal cell carcinoma (ccRCC). Using ccRCC RNA sequencing data from The Cancer Genome Atlas, we found that epithelial splicing regulatory protein 2 (ESRP2), one of the key regulators of alternative splicing in epithelial cells, is expressed in ccRCC. ESRP2 mRNA expression did not correlate with the overall survival rate of ccRCC patients, but the expression of some ESRP-target exons correlated with the good prognosis and with the expression of Arkadia (also known as RNF111) in ccRCC. Arkadia physically interacted with ESRP2, induced polyubiquitination and modulated its splicing function. Arkadia and ESRP2 suppressed ccRCC tumor growth in a coordinated manner. Lower expression of Arkadia correlated with advanced tumor stages and poor outcomes in ccRCC patients. This study thus reveals a novel tumor-suppressive role of the Arkadia-ESRP2 axis in ccRCC.
Collapse
|
30
|
Kumar S, Torres MP, Kaur S, Rachagani S, Joshi S, Johansson SL, Momi N, Baine MJ, Gilling CE, Smith LM, Wyatt TA, Jain M, Joshi SS, Batra SK. Smoking accelerates pancreatic cancer progression by promoting differentiation of MDSCs and inducing HB-EGF expression in macrophages. Oncogene 2015; 34:2052-60. [PMID: 24909166 PMCID: PMC4457293 DOI: 10.1038/onc.2014.154] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 04/17/2014] [Accepted: 04/21/2014] [Indexed: 12/22/2022]
Abstract
Smoking is an established risk factor for pancreatic cancer (PC), but late diagnosis limits the evaluation of its mechanistic role in the progression of PC. We used a well-established genetically engineered mouse model (LSL-K-ras(G12D)) of PC to elucidate the role of smoking during initiation and development of pancreatic intraepithelial neoplasia (PanIN). The 10-week-old floxed mice (K-ras(G12D); Pdx-1cre) and their control unfloxed (LSL-K-ras(G12D)) littermates were exposed to cigarette smoke (total suspended particles: 150 mg/m(3)) for 20 weeks. Smoke exposure significantly accelerated the development of PanIN lesions in the floxed mice, which correlated with tenfold increase in the expression of cytokeratin19. The systemic accumulation of myeloid-derived suppressor cells (MDSCs) decreased significantly in floxed mice compared with unfloxed controls (P<0.01) after the smoke exposure with the concurrent increase in the macrophage (P<0.05) and dendritic cell (DCs) (P<0.01) population. Further, smoking-induced inflammation (IFN-γ, CXCL2; P<0.05) was accompanied by enhanced activation of pancreatic stellate cells and elevated levels of serum retinoic acid-binding protein 4, indicating increased bioavailability of retinoic acid which contributes to differentiation of MDSCs to tumor-associated macrophages (TAMs) and DCs. TAMs predominantly contribute to the increased expression of heparin-binding epidermal growth factor-like growth factor (EGFR ligand) in pre-neoplastic lesions in smoke-exposed floxed mice that facilitate acinar-to-ductal metaplasia (ADM). Further, smoke exposure also resulted in partial suppression of the immune system early during PC progression. Overall, the present study provides a novel mechanism of smoking-induced increase in ADM in the presence of constitutively active K-ras mutation.
Collapse
Affiliation(s)
- S Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - MP Torres
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Joshi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - SL Johansson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - N Momi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - MJ Baine
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - CE Gilling
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - LM Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - TA Wyatt
- Division of Pulmonary, Critical Care, Sleep & Allergy, University of Nebraska Medical Center, Omaha, NE, USA
| | - M Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical center, Omaha, NE, USA
| | - SS Joshi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - SK Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical center, Omaha, NE, USA
| |
Collapse
|
31
|
DeCant BT, Principe DR, Guerra C, Pasca di Magliano M, Grippo PJ. Utilizing past and present mouse systems to engineer more relevant pancreatic cancer models. Front Physiol 2014; 5:464. [PMID: 25538623 PMCID: PMC4255505 DOI: 10.3389/fphys.2014.00464] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/11/2014] [Indexed: 12/14/2022] Open
Abstract
The study of pancreatic cancer has prompted the development of numerous mouse models that aim to recapitulate the phenotypic and mechanistic features of this deadly malignancy. This review accomplishes two tasks. First, it provides an overview of the models that have been used as representations of both the neoplastic and carcinoma phenotypes. Second, it presents new modeling schemes that ultimately will serve to more faithfully capture the temporal and spatial progression of the human disease, providing platforms for improved understanding of the role of non-epithelial compartments in disease etiology as well as evaluating therapeutic approaches.
Collapse
Affiliation(s)
- Brian T DeCant
- Department of Medicine, University of Illinois at Chicago Chicago, IL, USA
| | - Daniel R Principe
- Department of Medicine, University of Illinois at Chicago Chicago, IL, USA
| | - Carmen Guerra
- Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas Madrid, Spain
| | | | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago Chicago, IL, USA
| |
Collapse
|
32
|
Liou GY, Döppler H, Necela B, Edenfield B, Zhang L, Dawson DW, Storz P. Mutant KRAS-induced expression of ICAM-1 in pancreatic acinar cells causes attraction of macrophages to expedite the formation of precancerous lesions. Cancer Discov 2014; 5:52-63. [PMID: 25361845 DOI: 10.1158/2159-8290.cd-14-0474] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
UNLABELLED Desmoplasia and an inflammatory environment are defining features of pancreatic cancer. Unclear is how pancreatic cells that undergo oncogenic transformation can cross-talk with immune cells and how this contributes to the development of pancreatic lesions. Here, we demonstrate that pancreatic acinar cells expressing mutant KRAS can expedite their transformation to a duct-like phenotype by inducing local inflammation. Specifically, we show that KRAS(G12D) induces the expression of intercellular adhesion molecule-1 (ICAM-1), which serves as chemoattractant for macrophages. Infiltrating macrophages amplify the formation of KRAS(G12D)-caused abnormal pancreatic structures by remodeling the extracellular matrix and providing cytokines such as TNF. Depletion of macrophages or treatment with a neutralizing antibody for ICAM-1 in mice expressing oncogenic Kras under an acinar cell-specific promoter resulted in both a decreased formation of abnormal structures and decreased progression of acinar-to-ductal metaplasia to pancreatic intraepithelial neoplastic lesions. SIGNIFICANCE We here show that oncogenic KRAS in pancreatic acinar cells upregulates the expression of ICAM-1 to attract macrophages. Hence, our results reveal a direct cooperative mechanism between oncogenic Kras mutations and the inflammatory environment to drive the initiation of pancreatic cancer.
Collapse
Affiliation(s)
- Geou-Yarh Liou
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Heike Döppler
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Brian Necela
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Brandy Edenfield
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Lizhi Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - David W Dawson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
33
|
Lin CY, Chen HJ, Huang CC, Lai LC, Lu TP, Tseng GC, Kuo TT, Kuok QY, Hsu JL, Sung SY, Hung MC, Sher YP. ADAM9 promotes lung cancer metastases to brain by a plasminogen activator-based pathway. Cancer Res 2014; 74:5229-43. [PMID: 25060522 DOI: 10.1158/0008-5472.can-13-2995] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The transmembrane cell adhesion protein ADAM9 has been implicated in cancer cell migration and lung cancer metastasis to the brain, but the underpinning mechanisms are unclear and clinical support has been lacking. Here, we demonstrate that ADAM9 enhances the ability of tissue plasminogen activator (tPA) to cleave and stimulate the function of the promigratory protein CDCP1 to promote lung metastasis. Blocking this mechanism of cancer cell migration prolonged survival in tumor-bearing mice and cooperated with dexamethasone and dasatinib (a dual Src/Abl kinase inhibitor) treatment to enhance cytotoxic treatment. In clinical specimens, high levels of ADAM9 and CDCP1 correlated with poor prognosis and high risk of mortality in patients with lung cancer. Moreover, ADAM9 levels in brain metastases derived from lung tumors were relatively higher than the levels observed in primary lung tumors. Our results show how ADAM9 regulates lung cancer metastasis to the brain by facilitating the tPA-mediated cleavage of CDCP1, with potential implications to target this network as a strategy to prevent or treat brain metastatic disease.
Collapse
Affiliation(s)
- Chen-Yuan Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan. Division of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan
| | - Hung-Jen Chen
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chung Huang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Liang-Chuan Lai
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Tzu-Pin Lu
- YongLin Biomedical Engineering Center, National Taiwan University, Taipei, Taiwan
| | - Guan-Chin Tseng
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Ting-Ting Kuo
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Qian-Yu Kuok
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jennifer L Hsu
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan. Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Shian-Ying Sung
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Mien-Chie Hung
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan. Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Biotechnology, Asia University, Taichung, Taiwan. Graduate Institute for Cancer Biology, China Medical University, Taichung, Taiwan.
| | - Yuh-Pyng Sher
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan. Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
34
|
Abstract
Intratumoral hypoxia is a common feature of solid tumors. Recent advances in cancer biology indicate that hypoxia is not only a consequence of unrestrained tumor growth, but also plays an active role in promoting tumor progression, malignancy, and resistance to therapy. Hypoxia signaling is mediated by the hypoxia-inducible factors (HIFs), which are not only stabilized under hypoxia, but also by activated oncogenes or inactivated tumor suppressors under normoxia. Hypoxia is a prominent feature of the tumor microenvironment of pancreatic tumors, also characterized by the presence of a fibrotic reaction that promotes, and is also modulated by, hypoxia. As the mechanisms by which hypoxia signaling impacts invasion and metastasis in pancreatic cancer are being elucidated, hypoxia is emerging as a key determinant of pancreatic cancer malignancy as well as an important target for therapy. Herein we present an overview of recent advances in the understanding of the impact that hypoxia has in pancreatic cancer invasion and metastasis.
Collapse
Affiliation(s)
- Angela Yuen
- Tumor Microenvironment and Metastasis Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Begoña Díaz
- Tumor Microenvironment and Metastasis Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
35
|
Loss of HNF6 expression correlates with human pancreatic cancer progression. J Transl Med 2014; 94:517-27. [PMID: 24638272 PMCID: PMC4068339 DOI: 10.1038/labinvest.2014.47] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 01/28/2013] [Accepted: 02/10/2014] [Indexed: 01/27/2023] Open
Abstract
Normal pancreatic epithelium progresses through various stages of pancreatic intraepithelial neoplasms (PanINs) in the development of pancreatic ductal adenocarcinoma (PDAC). Transcriptional regulation of this progression is poorly understood. In mouse, the hepatic nuclear factor 6 (Hnf6) transcription factor is expressed in ductal cells and at lower levels in acinar cells of the adult pancreas, but not in mature endocrine cells. Hnf6 is critical for terminal differentiation of the ductal epithelium during embryonic development and for pancreatic endocrine cell specification. We previously showed that, in mice, loss of Hnf6 from the pancreatic epithelium during organogenesis results in increased duct proliferation and altered duct architecture, increased periductal fibrosis and acinar-to-ductal metaplasia. Here we show that decreased expression of HNF6 is strongly correlated with increased severity of PanIN lesions in samples of human pancreata and is absent from >90% of PDAC. Mouse models in which cancer progression can be analyzed from the earliest stages that are seldom accessible in humans support a role for Hnf6 loss in progression from early- to late-stage PanIN and PDAC. In addition, gene expression analyses of human pancreatic cancer reveal decreased expression of HNF6 and its direct and indirect target genes compared with normal tissue and upregulation of genes that act in opposition to HNF6 and its targets. The negative correlation between HNF6 expression and pancreatic cancer progression suggests that HNF6 maintains pancreatic epithelial homeostasis in humans, and that its loss contributes to the progression from PanIN to ductal adenocarcinoma. Insight on the role of HNF6 in pancreatic cancer development could lead to its use as a biomarker for early detection and prognosis.
Collapse
|
36
|
Shi C, Washington MK, Chaturvedi R, Drosos Y, Revetta FL, Weaver CJ, Buzhardt E, Yull FE, Blackwell TS, Sosa-Pineda B, Whitehead RH, Beauchamp RD, Wilson KT, Means AL. Fibrogenesis in pancreatic cancer is a dynamic process regulated by macrophage-stellate cell interaction. J Transl Med 2014; 94:409-21. [PMID: 24535260 PMCID: PMC3992484 DOI: 10.1038/labinvest.2014.10] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/15/2014] [Indexed: 12/31/2022] Open
Abstract
Pancreatic cancer occurs in the setting of a profound fibrotic microenvironment that often dwarfs the actual tumor. Although pancreatic fibrosis has been well studied in chronic pancreatitis, its development in pancreatic cancer is much less well understood. This article describes the dynamic remodeling that occurs from pancreatic precursors (pancreatic intraepithelial neoplasias (PanINs)) to pancreatic ductal adenocarcinoma, highlighting similarities and differences between benign and malignant disease. Although collagen matrix is a commonality throughout this process, early stage PanINs are virtually free of periostin while late stage PanIN and pancreatic cancer are surrounded by an increasing abundance of this extracellular matrix protein. Myofibroblasts also become increasingly abundant during progression from PanIN to cancer. From the earliest stages of fibrogenesis, macrophages are associated with this ongoing process. In vitro co-culture indicates there is cross-regulation between macrophages and pancreatic stellate cells (PaSCs), precursors to at least some of the fibrotic cell populations. When quiescent PaSCs were co-cultured with macrophage cell lines, the stellate cells became activated and the macrophages increased cytokine production. In summary, fibrosis in pancreatic cancer involves a complex interplay of cells and matrices that regulate not only the tumor epithelium but the composition of the microenvironment itself.
Collapse
Affiliation(s)
- Chanjuan Shi
- Dept. of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville TN
| | - M. Kay Washington
- Dept. of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville TN
| | | | - Yiannis Drosos
- Dept. of Genetics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Frank L. Revetta
- Dept. of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville TN
| | | | | | - Fiona E. Yull
- Dept. of Cancer Biology, Vanderbilt University, Nashville TN
| | | | | | | | - R. Daniel Beauchamp
- Dept. of Surgery, Vanderbilt University, Nashville TN,Dept. of Cell and Developmental Biology, Vanderbilt University, Nashville TN
| | - Keith T. Wilson
- Dept. of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville TN,Dept. of Medicine, Vanderbilt University, Nashville TN,Dept. of Cancer Biology, Vanderbilt University, Nashville TN,Veterans Affairs Tennessee Valley Healthcare System, Nashville TN
| | - Anna L. Means
- Dept. of Surgery, Vanderbilt University, Nashville TN,Dept. of Cell and Developmental Biology, Vanderbilt University, Nashville TN
| |
Collapse
|
37
|
Bailey JM, DelGiorno KE, Crawford HC. The secret origins and surprising fates of pancreas tumors. Carcinogenesis 2014; 35:1436-40. [PMID: 24583923 DOI: 10.1093/carcin/bgu056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is especially deadly due to its recalcitrance to current therapies. One of the unique qualities of PDA that may contribute to this resistance is a striking plasticity of differentiation states starting at tumor formation and continuing throughout tumor progression, including metastasis. Here, we explore the earliest steps of tumor formation and neoplastic progression and how this results in a fascinating cellular heterogeneity that is probably critical for tumor survival and progression. We hypothesize that reinforcing differentiation pathways run awry or targeting morphologically and molecularly distinct tumor stem-like cells may hold promise for future treatments of this deadly disease.
Collapse
Affiliation(s)
- Jennifer M Bailey
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA, Department of Medicine, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA and Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL 32224, USA
| | - Kathleen E DelGiorno
- Department of Medicine, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA and
| | - Howard C Crawford
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL 32224, USA
| |
Collapse
|
38
|
Melanoma genotypes and phenotypes get personal. J Transl Med 2013; 93:858-67. [PMID: 23817084 DOI: 10.1038/labinvest.2013.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 05/28/2013] [Accepted: 06/01/2013] [Indexed: 01/23/2023] Open
Abstract
Traditionally, the diagnosis of metastatic melanoma was terminal to most patients. However, the advancements towards understanding the fundamental etiology, pathophysiology, and treatment have raised melanoma to the forefront of contemporary medicine. Indeed, the evidence of durable remissions are being heard ever more frequently in clinics around the globe. Despite having more gene mutations per cell than any other type of cancer, investigators are overcoming complex genomic landscapes, signaling pathways, and immune checkpoints by generating novel technological methods and clinical protocols with breath-taking speed. Significant progress in deciphering molecular genetics, epigenetics, kinase-driven networks, metabolomics, and immune-enhancing pathways to achieve personalized and positive outcomes has truly provided new hope for melanoma patients. However, obstacles requiring breakthroughs include understanding the influence of sunlight exposure on melanoma etiology, and overcoming all too frequently acquired drug resistance, complicating targeted therapy. Pathologists continue to have critically important roles in advancing the field, particularly in the area of transitioning from microscope-based diagnostic reports to pharmacogenomics through molecularly informed tumor boards. Although melanoma is no longer considered just 'one disease', pathologists will continue this rapidly progressing and exciting journey to identify tumor subtypes, to utilize tumorgraft or so-called patient-derived xenograft (PDX) models, and to develop companion diagnostics to keep pace with the bewildering breakthroughs occurring on a regular basis. Exactly which combination of drugs will ultimately be required to eradicate melanoma cells remains to be determined. However, it is clear that pathologists who are as dedicated to melanoma as the pioneering pathologist Dr Sidney Farber was committed to childhood cancers, will be required as the battle against melanoma continues. In this review, we describe what sets melanoma apart from other tumors, and demonstrate how lessons learned in the melanoma clinic are being transferred to many other types of aggressive neoplasms.
Collapse
|