1
|
Kanzaki R, Reid S, Bolivar P, Sjölund J, Staaf J, Larsson S, Shintani Y, Pietras K. FHL2 expression by cancer-associated fibroblasts promotes metastasis and angiogenesis in lung adenocarcinoma. Int J Cancer 2025; 156:431-446. [PMID: 39244734 DOI: 10.1002/ijc.35174] [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: 09/15/2023] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024]
Abstract
Cancer-associated fibroblasts (CAFs) contribute to the progression of lung cancer. Four and a half LIM domain protein-2 (FHL2) is a component of focal adhesion structures. We analyzed the function of FHL2 expressed by CAFs in lung adenocarcinoma. Expression of FHL2 in fibroblast subtypes was investigated using database of single-cell RNA-sequencing of lung cancer tissue. The role of FHL2 in the proliferation and migration of CAFs was assessed. The effects of FHL2 knockout on the migration and invasion of human lung adenocarcinoma cells and tube formation of endothelial cells induced by CAF-conditioned medium (CM) were evaluated. The effect of FHL2 knockout in CAFs on metastasis was determined using a murine orthotopic lung cancer model. The prognostic significance of stromal FHL2 was assessed by immunohistochemistry in human adenocarcinoma specimens. FHL2 is highly expressed in myofibroblasts in cancer tissue. TGF-β1 upregulated FHL2 expression in CAFs and FHL2 knockdown attenuated CAF proliferation. FHL2 knockout reduced CAF induced migration of A110L and H23 human lung adenocarcinoma cell lines, and the induction of tube formation of endothelial cells. FHL2 knockout reduced CAF-induced metastasis of lung adenocarcinomas in an orthotopic model in vivo. The concentration of Osteopontin (OPN) in CM from CAF was downregulated by FHL2 knockout. siRNA silencing and antibody blocking of OPN reduced the pro-migratory effect of CM from CAF on lung cancer cells. In resected lung adenocarcinoma specimens, positive stromal FHL2 expression was significantly associated with higher microvascular density and worse prognosis. In conclusion, FHL2 expression by CAFs enhances the progression of lung adenocarcinoma by promoting angiogenesis and metastasis.
Collapse
Affiliation(s)
- Ryu Kanzaki
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of General Thoracic Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Steven Reid
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Paulina Bolivar
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Johan Staaf
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Sara Larsson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University Cancer Centre, Lund University, Lund, Sweden
| |
Collapse
|
2
|
Chang C, Tang X, Schönthal AH, Chen M, Woodley DT, Wang Y, Liang C, Li W. Discovery of Cell Number-Interstitial Fluid Volume (CIF) Ratio Reveals Secretory Autophagy Pathway to Supply eHsp90α for Wound Healing. Cells 2024; 13:1280. [PMID: 39120311 PMCID: PMC11312289 DOI: 10.3390/cells13151280] [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: 06/11/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Cell secretion repairs tissue damage and restores homeostasis throughout adult life. The extracellular heat shock protein-90alpha (eHsp90α) has been reported as an exosome cargo and a potential driver of wound healing. However, neither the mechanism of secretion nor the genetic evidence for eHsp90α in wound healing has been substantiated. Herein, we show that tissue injury causes massive deposition of eHsp90α in tissues and secretion of eHsp90α by cells. Sequential centrifugations of conditioned medium from relevant cell lines revealed the relative distributions of eHsp90α in microvesicle, exosome and trypsin-sensitive supernatant fractions to be approximately <2%, <4% and >95%, respectively. Establishing the cell-number-to-interstitial-fluid-volume (CIF) ratio for the microenvironment of human tissues as 1 × 109 cells: 1 mL interstitial fluid enabled us to predict the corresponding tissue concentrations of eHsp90α in these fractions as 3.74 μg/mL, 5.61 μg/mL and 178 μg/mL. Remarkably, the 178 μg/mL eHsp90α matches the previously reported 100-300 μg/mL of recombinant eHsp90α whose topical application promotes maximum wound healing in animal models. More importantly, we demonstrate that two parallel secretory autophagy-regulating gene families, the autophagy-regulating (AR) genes and the Golgi reassembly-stacking protein (GRASP) genes work together to mediate the secretion of the physiological concentration of eHsp90α to promote wound healing. Thus, utilization of the CIF ratio-based extrapolation method may enable investigators to rapidly predict biomarker targets from cell-conditioned-medium data.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Dermatology and the USC-Norris Comprehensive Cancer Centre, Los Angeles, CA 90089, USA; (C.C.); (X.T.); (M.C.); (D.T.W.)
| | - Xin Tang
- Department of Dermatology and the USC-Norris Comprehensive Cancer Centre, Los Angeles, CA 90089, USA; (C.C.); (X.T.); (M.C.); (D.T.W.)
| | - Axel H. Schönthal
- Department of Molecular Microbiology & Immunology, University of Southern California Keck Medical Centre, Los Angeles, CA 90033, USA;
| | - Mei Chen
- Department of Dermatology and the USC-Norris Comprehensive Cancer Centre, Los Angeles, CA 90089, USA; (C.C.); (X.T.); (M.C.); (D.T.W.)
| | - David T. Woodley
- Department of Dermatology and the USC-Norris Comprehensive Cancer Centre, Los Angeles, CA 90089, USA; (C.C.); (X.T.); (M.C.); (D.T.W.)
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI 48109, USA;
| | - Chengyu Liang
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA;
| | - Wei Li
- Department of Dermatology and the USC-Norris Comprehensive Cancer Centre, Los Angeles, CA 90089, USA; (C.C.); (X.T.); (M.C.); (D.T.W.)
| |
Collapse
|
3
|
Li P, Cui P, Yue Q, Xu Z, Liu Z. Exploring the potential biological significance of KDELR family genes in lung adenocarcinoma. Sci Rep 2024; 14:14820. [PMID: 38937522 PMCID: PMC11211404 DOI: 10.1038/s41598-024-65425-2] [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: 01/24/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
The Lys-Asp-Glu-Leu receptor (KDELR) family genes play critical roles in a variety of biological processes in different tumors. Our study aimed to provide a comprehensive analysis of the potential roles of KDELRs in lung adenocarcinoma (LUAD). Utilizing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, as well as clinical samples, we conducted a series of analyses and validations using R software tools and various online resources. The results showed that KDELR family genes and proteins were highly expressed and associated with a poor prognosis of LUAD. Promoter hypomethylation and the competing endogenous RNA (ceRNA) network of PCAT6/hsa-miR-326/KDELR1 might be potential causes of aberrant KDELR1 overexpression in LUAD. Three key Transcription factors (TFs) (SPI1, EP300, and MAZ) and a TFs-miRNAs-KDELRs network (involving 11 TFs) might be involved in modulating KDELRs expression abnormalities. Gene Set Enrichment Analysis (GSEA) indicated enrichment of genes highly expressing KDELR1, KDELR2, and KDELR3 in MTORC1_SIGNALING, P53_PATHWAY, and ANGIOGENESIS. Negative correlations between KDELRs expression and CD8 + T cell infiltration, as well as CTLA-4 expression. Our multiple analyses suggested that the KDELRs are important signaling molecules in LUAD. These results provided novel insights for developing prognostic markers and novel therapies of LUAD.
Collapse
Affiliation(s)
- Peitong Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Pengfei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Qing Yue
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zijun Xu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Ziling Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
4
|
Aluksanasuwan S, Somsuan K, Ngoenkam J, Chiangjong W, Rongjumnong A, Morchang A, Chutipongtanate S, Pongcharoen S. Knockdown of heat shock protein family D member 1 (HSPD1) in lung cancer cell altered secretome profile and cancer-associated fibroblast induction. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119736. [PMID: 38663552 DOI: 10.1016/j.bbamcr.2024.119736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/02/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The crosstalk between lung cancer cells and cancer-associated fibroblast (CAF) is pivotal in cancer progression. Heat shock protein family D member 1 (HSPD1) is a potential prognostic biomarker associated with the tumor microenvironment in lung adenocarcinoma (LUAD). However, the role of HSPD1 in CAF activation remains unclear. This study established stable HSPD1-knockdown A549 lung cancer cells using a lentivirus-mediated shRNA transduction. A targeted label-free proteomic analysis identified six significantly altered secretory proteins in the shHSPD1-A549 secretome compared to shControl-A549. Functional enrichment analysis highlighted their involvement in cell-to-cell communication and immune responses within the tumor microenvironment. Additionally, most altered proteins exhibited positive correlations and significant prognostic impacts on LUAD patient survival. Investigations on the effects of lung cancer secretomes on lung fibroblast WI-38 cells revealed that the shControl-A549 secretome stimulated fibroblast proliferation, migration, and CAF marker expression. These effects were reversed upon the knockdown of HSPD1 in A549 cells. Altogether, our findings illustrate the role of HSPD1 in mediating CAF induction through secretory proteins, potentially contributing to the progression and aggressiveness of lung cancer.
Collapse
Affiliation(s)
- Siripat Aluksanasuwan
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand.
| | - Keerakarn Somsuan
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Wararat Chiangjong
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Artitaya Rongjumnong
- Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Atthapan Morchang
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Somchai Chutipongtanate
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0056, USA
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand.
| |
Collapse
|
5
|
Korbecki J, Bosiacki M, Szatkowska I, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. The Clinical Significance and Involvement in Molecular Cancer Processes of Chemokine CXCL1 in Selected Tumors. Int J Mol Sci 2024; 25:4365. [PMID: 38673949 PMCID: PMC11050300 DOI: 10.3390/ijms25084365] [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: 01/29/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Chemokines play a key role in cancer processes, with CXCL1 being a well-studied example. Due to the lack of a complete summary of CXCL1's role in cancer in the literature, in this study, we examine the significance of CXCL1 in various cancers such as bladder, glioblastoma, hemangioendothelioma, leukemias, Kaposi's sarcoma, lung, osteosarcoma, renal, and skin cancers (malignant melanoma, basal cell carcinoma, and squamous cell carcinoma), along with thyroid cancer. We focus on understanding how CXCL1 is involved in the cancer processes of these specific types of tumors. We look at how CXCL1 affects cancer cells, including their proliferation, migration, EMT, and metastasis. We also explore how CXCL1 influences other cells connected to tumors, like promoting angiogenesis, recruiting neutrophils, and affecting immune cell functions. Additionally, we discuss the clinical aspects by exploring how CXCL1 levels relate to cancer staging, lymph node metastasis, patient outcomes, chemoresistance, and radioresistance.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Iwona Szatkowska
- Department of Ruminants Science, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology, Klemensa Janickiego 29 St., 71-270 Szczecin, Poland;
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| |
Collapse
|
6
|
Selvaraj C, Panwar U, Ramalingam KR, Vijayakumar R, Singh SK. Exploring the macromolecules for secretory pathway in cancer disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:55-83. [PMID: 36707206 DOI: 10.1016/bs.apcsb.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Secretory proteins play an important role in the tumor microenvironment and are widely distributed throughout tumor tissues. Tumor cells secrete a protein that mediates communication between tumor cells and stromal cells, thereby controlling tumor growth and affecting the success of cancer treatments in the clinic. The cancer secretome is produced by various secretory pathways and has a wide range of applications in oncoproteomics. Secretory proteins are involved in cancer development and tumor cell migration, and thus serve as biomarkers or effective therapeutic targets for a variety of cancers. Several proteomic strategies have recently been used for the analysis of cancer secretomes in order to gain a better understanding and elaborate interpretation. For instance, the development of exosome proteomics, degradomics, and tumor-host cell interaction provide clear information regarding the mechanism of cancer pathobiology. In this chapter, we emphasize the recent advances in secretory protein and the challenges in the field of secretome analysis and their clinical applications.
Collapse
Affiliation(s)
- Chandrabose Selvaraj
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India.
| | - Umesh Panwar
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Karthik Raja Ramalingam
- Department of Biotechnology, Division of Research and Innovation, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Rajendran Vijayakumar
- Department of Biology, College of Science in Zulfi, Majmaah University, Majmaah, Saudi Arabia
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
7
|
Rix LLR, Sumi NJ, Hu Q, Desai B, Bryant AT, Li X, Welsh EA, Fang B, Kinose F, Kuenzi BM, Chen YA, Antonia SJ, Lovly CM, Koomen JM, Haura EB, Marusyk A, Rix U. IGF-binding proteins secreted by cancer-associated fibroblasts induce context-dependent drug sensitization of lung cancer cells. Sci Signal 2022; 15:eabj5879. [PMID: 35973030 PMCID: PMC9528501 DOI: 10.1126/scisignal.abj5879] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cancer-associated fibroblasts (CAFs) in the tumor microenvironment are often linked to drug resistance. Here, we found that coculture with CAFs or culture in CAF-conditioned medium unexpectedly induced drug sensitivity in certain lung cancer cell lines. Gene expression and secretome analyses of CAFs and normal lung-associated fibroblasts (NAFs) revealed differential abundance of insulin-like growth factors (IGFs) and IGF-binding proteins (IGFBPs), which promoted or inhibited, respectively, signaling by the receptor IGF1R and the kinase FAK. Similar drug sensitization was seen in gefitinib-resistant, EGFR-mutant PC9GR lung cancer cells treated with recombinant IGFBPs. Conversely, drug sensitivity was decreased by recombinant IGFs or conditioned medium from CAFs in which IGFBP5 or IGFBP6 was silenced. Phosphoproteomics and receptor tyrosine kinase (RTK) array analyses indicated that exposure of PC9GR cells to CAF-conditioned medium also inhibited compensatory IGF1R and FAK signaling induced by the EGFR inhibitor osimertinib. Combined small-molecule inhibition of IGF1R and FAK phenocopied the CAF-mediated effects in culture and increased the antitumor effect of osimertinib in mice. Cells that were osimertinib resistant and had MET amplification or showed epithelial-to-mesenchymal transition also displayed residual sensitivity to IGFBPs. Thus, CAFs promote or reduce drug resistance in a context-dependent manner, and deciphering the relationship between the differential content of CAF secretomes and the signaling dependencies of the tumor may reveal effective combination treatment strategies.
Collapse
Affiliation(s)
- Lily L. Remsing Rix
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA
| | - Natalia J. Sumi
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620, USA
| | - Qianqian Hu
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620, USA
| | - Bina Desai
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620, USA
| | - Annamarie T. Bryant
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA
| | - Xueli Li
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA
| | - Eric A. Welsh
- Biostatistics and Bioinformatics Shared Resource, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Bin Fang
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Brent M. Kuenzi
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA.,Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620, USA
| | - Y. Ann Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Scott J. Antonia
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Christine M. Lovly
- Department of Medicine, Vanderbilt University Medical Center; Nashville, TN 37232, USA
| | - John M. Koomen
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA,Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Andriy Marusyk
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA,Department of Cancer Physiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Uwe Rix
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, Florida 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA,Corresponding author.
| |
Collapse
|
8
|
Zhang S, Wang C, Ju J, Wang C. Extracellular Hsp90α Supports the ePKM2-GRP78-AKT Axis to Promote Tumor Metastasis. Front Oncol 2022; 12:906080. [PMID: 35847880 PMCID: PMC9280132 DOI: 10.3389/fonc.2022.906080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor-secreted proteins can provide numerous molecular targets for cancer diagnosis and treatment. Of note, pyruvate kinase M2 (PKM2) is secreted by tumor cells to promote malignant progression, while its regulatory mechanism or the interacting network remains uncovered. In the present study, we identified extracellular heat shock protein 90 alpha (eHsp90α) as one potential interacting protein of ePKM2 by mass spectrometry (MS), which was further verified by pull-down and co-immunoprecipitation analysis. Later, we found that eHsp90α enhanced the effect of ePKM2 on migration and invasion of lung cancer cells. Blocking of Hsp90α activity, on the other hand, attenuated tumor migration or invasion induced by ePKM2. Eventually, the in vivo role of Hsp90α in regulating ePKM2 activity was validated by the mouse xenograft tumor model. Mechanistically, we found that eHsp90α binds to and stabilizes ePKM2 to protect it from degradation in the extracellular environment. Besides, eHsp90α promoted the interaction of ePKM2 with cell surface receptor GRP78, which leads to the activation of the ePKM2/GRP78/AKT axis. Collectively, we unraveled the novel molecular mechanism of eHsp90α in regulating ePKM2 activity during tumor progression, which is beneficial for the development of new treatments against lung cancer.
Collapse
Affiliation(s)
- Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caihong Wang
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiujun Ju
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Caixia Wang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
- *Correspondence: Caixia Wang,
| |
Collapse
|
9
|
Huang M, Xiong D, Pan J, Zhang Q, Wang Y, Myers CR, Johnson BD, Hardy M, Kalyanaraman B, You M. Prevention of Tumor Growth and Dissemination by In Situ Vaccination with Mitochondria-Targeted Atovaquone. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2101267. [PMID: 35243806 PMCID: PMC9036031 DOI: 10.1002/advs.202101267] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 02/09/2022] [Indexed: 05/06/2023]
Abstract
Atovaquone, an FDA-approved drug for malaria, is known to inhibit mitochondrial electron transport. A recently synthesized mitochondria-targeted atovaquone increased mitochondrial accumulation and antitumor activity in vitro. Using an in situ vaccination approach, local injection of mitochondria-targeted atovaquone into primary tumors triggered potent T cell immune responses locally and in distant tumor sites. Mitochondria-targeted atovaquone treatment led to significant reductions of both granulocytic myeloid-derived suppressor cells and regulatory T cells in the tumor microenvironment. Mitochondria-targeted atovaquone treatment blocks the expression of genes involved in oxidative phosphorylation and glycolysis in granulocytic-myeloid-derived suppressor cells and regulatory T cells, which may lead to death of granulocytic-myeloid-derived suppressor cells and regulatory T cells. Mitochondria-targeted atovaquone inhibits expression of genes for mitochondrial complex components, oxidative phosphorylation, and glycolysis in both granulocytic-myeloid-derived suppressor cells and regulatory T cells. The resulting decreases in intratumoral granulocytic-myeloid-derived suppressor cells and regulatory T cells could facilitate the observed increase in tumor-infiltrating CD4+ T cells. Mitochondria-targeted atovaquone also improves the anti-tumor activity of PD-1 blockade immunotherapy. The results implicate granulocytic-myeloid-derived suppressor cells and regulatory T cells as novel targets of mitochondria-targeted atovaquone that facilitate its antitumor efficacy.
Collapse
Affiliation(s)
- Mofei Huang
- Center for Cancer Prevention, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Donghai Xiong
- Center for Cancer Prevention, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Jing Pan
- Center for Cancer Prevention, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Qi Zhang
- Center for Cancer Prevention, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Yian Wang
- Center for Cancer Prevention, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Charles R Myers
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Bryon D Johnson
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Micael Hardy
- Aix Marseille Univ, CNRS, ICR, UMR 7273, Marseille, 13013, France
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Ming You
- Center for Cancer Prevention, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| |
Collapse
|
10
|
Abstract
Cachexia, a wasting syndrome that is often associated with cancer, is one of the primary causes of death in cancer patients. Cancer cachexia occurs largely due to systemic metabolic alterations stimulated by tumors. Despite the prevalence of cachexia, our understanding of how tumors interact with host tissues and how they affect metabolism is limited. Among the challenges of studying tumor-host tissue crosstalk are the complexity of cancer itself and our insufficient knowledge of the factors that tumors release into the blood. Drosophila is emerging as a powerful model in which to identify tumor-derived factors that influence systemic metabolism and tissue wasting. Strikingly, studies that are characterizing factors derived from different fly tumor cachexia models are identifying both common and distinct cachectic molecules, suggesting that cachexia is more than one disease and that fly models can help identify these differences. Here, we review what has been learned from studies of tumor-induced organ wasting in Drosophila and discuss the open questions.
Collapse
Affiliation(s)
- Ying Liu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro Saavedra
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
| |
Collapse
|
11
|
Sunaga N, Miura Y, Kasahara N, Sakurai R. Targeting Oncogenic KRAS in Non-Small-Cell Lung Cancer. Cancers (Basel) 2021; 13:cancers13235956. [PMID: 34885068 PMCID: PMC8656763 DOI: 10.3390/cancers13235956] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary v-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS) is the most common driver in NSCLC, and targeting oncogenic KRAS is a major challenge in the treatment of non-small-cell lung cancer (NSCLC). While several covalent KRAS G12C inhibitors have emerged as a novel anti-KRAS therapy, the development of combined therapies involving the targeting of oncogenic KRAS plus other targeted drugs is still required given the vast heterogeneity of KRAS-mutated tumors. In this review, we summarize the biological and immunological characteristics of oncogenic KRAS-driven NSCLC and the preclinical and clinical evidence for mutant KRAS-targeted therapies. We also discuss the mechanisms of resistance to KRAS G12C inhibitors and possible therapeutic strategies to overcome this drug resistance. Abstract Recent advances in molecular biology and the resultant identification of driver oncogenes have achieved major progress in precision medicine for non-small-cell lung cancer (NSCLC). v-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS) is the most common driver in NSCLC, and targeting KRAS is considerably important. The recent discovery of covalent KRAS G12C inhibitors offers hope for improving the prognosis of NSCLC patients, but the development of combination therapies corresponding to tumor characteristics is still required given the vast heterogeneity of KRAS-mutated NSCLC. In this review, we summarize the current understanding of KRAS mutations regarding the involvement of malignant transformation and describe the preclinical and clinical evidence for targeting KRAS-mutated NSCLC. We also discuss the mechanisms of resistance to KRAS G12C inhibitors and possible combination treatment strategies to overcome this drug resistance.
Collapse
Affiliation(s)
- Noriaki Sunaga
- Department of Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
- Correspondence: ; Tel.: +81-27-220-8000
| | - Yosuke Miura
- Department of Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
| | - Norimitsu Kasahara
- Innovative Medical Research Center, Gunma University Hospital, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
| | - Reiko Sakurai
- Oncology Center, Gunma University Hospital, 3-39-15 Showa-machi, Maebashi 371-8511, Gunma, Japan;
| |
Collapse
|
12
|
Potent inhibition of tumour cell proliferation and immunoregulatory function by mitochondria-targeted atovaquone. Sci Rep 2020; 10:17872. [PMID: 33087770 PMCID: PMC7578061 DOI: 10.1038/s41598-020-74808-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022] Open
Abstract
The FDA-approved prophylactic antimalarial drug atovaquone (ATO) recently was repurposed as an antitumor drug. Studies show that ATO exerts a profound antiproliferative effect in several cancer cells, including breast, ovarian, and glioma. Analogous to the mechanism of action proposed in parasites, ATO inhibits mitochondrial complex III and cell respiration. To enhance the chemotherapeutic efficacy and oxidative phosphorylation inhibition, we developed a mitochondria-targeted triphenylphosphonium-conjugated ATO with varying alkyl side chains (Mito4-ATO, Mito10-ATO, Mito12-ATO, and Mito16-ATO). Results show, for the first time, that triphenylphosphonium-conjugated ATO potently enhanced the antiproliferative effect of ATO in cancer cells and, depending upon the alkyl chain length, the molecular target of inhibition changes from mitochondrial complex III to complex I. Mito4-ATO and Mito10-ATO inhibit both pyruvate/malate-dependent complex I and duroquinol-dependent complex III-induced oxygen consumption whereas Mito12-ATO and Mito16-ATO inhibit only complex I-induced oxygen consumption. Mitochondrial target shifting may have immunoregulatory implications.
Collapse
|
13
|
Jäger B, Klatt D, Plappert L, Golpon H, Lienenklaus S, Barbosa PD, Schambach A, Prasse A. CXCR4/MIF axis amplifies tumor growth and epithelial-mesenchymal interaction in non-small cell lung cancer. Cell Signal 2020; 73:109672. [DOI: 10.1016/j.cellsig.2020.109672] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022]
|
14
|
Butera G, Brandi J, Cavallini C, Scarpa A, Lawlor RT, Scupoli MT, Marengo E, Cecconi D, Manfredi M, Donadelli M. The Mutant p53-Driven Secretome Has Oncogenic Functions in Pancreatic Ductal Adenocarcinoma Cells. Biomolecules 2020; 10:biom10060884. [PMID: 32526853 PMCID: PMC7356389 DOI: 10.3390/biom10060884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/30/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
The cancer secretome is a rich repository of useful information for both cancer biology and clinical oncology. A better understanding of cancer secretome is particularly relevant for pancreatic ductal adenocarcinoma (PDAC), whose extremely high mortality rate is mainly due to early metastasis, resistance to conventional treatments, lack of recognizable symptoms, and assays for early detection. TP53 gene is a master transcriptional regulator controlling several key cellular pathways and it is mutated in ~75% of PDACs. We report the functional effect of the hot-spot p53 mutant isoforms R175H and R273H on cancer cell secretome, showing their influence on proliferation, chemoresistance, apoptosis, and autophagy, as well as cell migration and epithelial-mesenchymal transition. We compared the secretome of p53-null AsPC-1 PDAC cells after ectopic over-expression of R175H-mutp53 or R273H-mutp53 to identify the differentially secreted proteins by mutant p53. By using high-resolution SWATH-MS technology, we found a great number of differentially secreted proteins by the two p53 mutants, 15 of which are common to both mutants. Most of these secreted proteins are reported to promote cancer progression and epithelial-mesenchymal transition and might constitute a biomarker secreted signature that is driven by the hot-spot p53 mutants in PDAC.
Collapse
Affiliation(s)
- Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (G.B.); (M.T.S.)
| | - Jessica Brandi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Chiara Cavallini
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, 37134 Verona, Italy;
| | - Aldo Scarpa
- Department of Diagnostics and Public health, Section of Pathology, University of Verona, 37134 Verona, Italy;
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Rita T. Lawlor
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (G.B.); (M.T.S.)
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, 37134 Verona, Italy;
| | - Emílio Marengo
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Italy, ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Italy, ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Italy, CAAD, corso Trieste 15/A, 28100 Novara, Italy
- Correspondence: (M.M.); (M.D.); Tel.: +39-032-1660810 (M.M.); +39-045-8027281 (M.D.); Fax: +39-045-8027170 (M.D.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (G.B.); (M.T.S.)
- Correspondence: (M.M.); (M.D.); Tel.: +39-032-1660810 (M.M.); +39-045-8027281 (M.D.); Fax: +39-045-8027170 (M.D.)
| |
Collapse
|
15
|
Wang C, Zhang S, Liu J, Tian Y, Ma B, Xu S, Fu Y, Luo Y. Secreted Pyruvate Kinase M2 Promotes Lung Cancer Metastasis through Activating the Integrin Beta1/FAK Signaling Pathway. Cell Rep 2020; 30:1780-1797.e6. [PMID: 32049010 DOI: 10.1016/j.celrep.2020.01.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/01/2019] [Accepted: 01/10/2020] [Indexed: 12/31/2022] Open
Abstract
Cancer cell-derived secretomes have been documented to play critical roles in cancer progression. Intriguingly, alternative extracellular roles of intracellular proteins are involved in various steps of tumor progression, which can offer strategies to fight cancer. Herein, we identify lung cancer progression-associated secretome signatures using mass spectrometry analysis. Among them, PKM2 is verified to be highly expressed and secreted in lung cancer cells and clinical samples. Functional analyses demonstrates that secreted PKM2 facilitates tumor metastasis. Furthermore, mass spectrometry analysis and functional validation identify integrin β1 as a receptor of secreted PKM2. Mechanistically, secreted PKM2 directly bound to integrin β1 and subsequently activated the FAK/SRC/ERK axis to promote tumor metastasis. Collectively, our findings suggest that PKM2 is a potential serum biomarker for diagnosing lung cancer and that targeting the secreted PKM2-integrin β1 axis can inhibit lung cancer development, which provides evidence of a potential therapeutic strategy in lung cancer.
Collapse
Affiliation(s)
- Caihong Wang
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
| | - Shaosen Zhang
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
| | - Jie Liu
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
| | - Yang Tian
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
| | - Boyuan Ma
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
| | - Siran Xu
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
| | - Yan Fu
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
| | - Yongzhang Luo
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China; The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
16
|
Gong Y, Wang C, Jiang Y, Zhang S, Feng S, Fu Y, Luo Y. Metformin Inhibits Tumor Metastasis through Suppressing Hsp90α Secretion in an AMPKα1-PKCγ Dependent Manner. Cells 2020; 9:cells9010144. [PMID: 31936169 PMCID: PMC7016760 DOI: 10.3390/cells9010144] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 12/27/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Metformin has been documented in epidemiological studies to mitigate tumor progression. Previous reports show that metformin inhibits tumor migration in several cell lines, such as MCF-7 and H1299, but the mechanisms whereby metformin exerts its inhibitory effects on tumor metastasis remain largely unknown. The secreted proteins in cancer cell-derived secretome have been reported to play important roles in tumor metastasis, but whether metformin has an effect on tumor secretome remains unclear. Here we show that metformin inhibits tumor metastasis by suppressing Hsp90α (heat shock protein 90α) secretion. Mass spectrometry (MS) analysis and functional validation identify that eHsp90α (extracellular Hsp90α) is one of the most important secreted proteins for metformin to inhibit tumor cells migration, invasion and metastasis both in vitro and in vivo. Moreover, we find that metformin inhibits Hsp90α secretion in an AMPKα1 dependent manner. Our data elucidate that AMPKα1 (AMP-activated protein kinase α1) decreases the phosphorylation level of Hsp90α by inhibiting the kinase activity of PKCγ (protein kinase Cγ), which suppresses the membrane translocation and secretion of Hsp90α. Collectively, our results illuminate that metformin inhibits tumor metastasis by suppressing Hsp90α secretion in an AMPKα1 dependent manner.
Collapse
Affiliation(s)
- Yuanchao Gong
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; (Y.G.); (C.W.); (Y.J.); (S.Z.); (S.F.); (Y.F.)
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Caihong Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; (Y.G.); (C.W.); (Y.J.); (S.Z.); (S.F.); (Y.F.)
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yi Jiang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; (Y.G.); (C.W.); (Y.J.); (S.Z.); (S.F.); (Y.F.)
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shaosen Zhang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; (Y.G.); (C.W.); (Y.J.); (S.Z.); (S.F.); (Y.F.)
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shi Feng
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; (Y.G.); (C.W.); (Y.J.); (S.Z.); (S.F.); (Y.F.)
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yan Fu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; (Y.G.); (C.W.); (Y.J.); (S.Z.); (S.F.); (Y.F.)
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yongzhang Luo
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing 100084, China; (Y.G.); (C.W.); (Y.J.); (S.Z.); (S.F.); (Y.F.)
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing 100084, China
- Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Correspondence: ; Tel.: +86-10-6277-2897; Fax: +86-10-6279-4691
| |
Collapse
|
17
|
Abstract
Mass spectrometry-based proteomics analysis could categorize proteins and study their interactions in large scale in human cancers. By this method, many proteins are upregulated or downregulated in esophageal squamous cell carcinoma (ESCC) when compared to nonneoplastic esophageal mucosae. The method can also be used to identify novel, effective biomarkers for early diagnosis or predict prognosis of patients with ESCC. These changes are associated with different clinical and pathological parameters. Different biological matrices such as pathological tissue, body fluids, and cancer cell lines-based proteomics have widely been used. Herein, we described cell line-based label-free shotgun proteomics (in-solution tryptic digestion) to identify the protein biomarkers differently expressed in ESCC.
Collapse
|
18
|
Pan J, Zhang Q, Palen K, Wang L, Qiao L, Johnson B, Sei S, Shoemaker RH, Lubet RA, Wang Y, You M. Potentiation of Kras peptide cancer vaccine by avasimibe, a cholesterol modulator. EBioMedicine 2019; 49:72-81. [PMID: 31680003 PMCID: PMC6945201 DOI: 10.1016/j.ebiom.2019.10.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 01/17/2023] Open
Abstract
Background No effective approaches to target mutant Kras have yet been developed. Immunoprevention using KRAS-specific antigenic peptides to trigger T cells capable of targeting tumor cells relies heavily on lipid metabolism. To facilitate better TCR/peptide/MHC interactions that result in better cancer preventive efficacy, we combined KVax with avasimibe, a specific ACAT1 inhibitor, tested their anti-cancer efficacy in mouse lung cancer models, where Kras mutation was induced before vaccination. Methods Control of tumor growth utilizing a multi-peptide Kras vaccine was tested in combination with avasimibe in a syngeneic lung cancer mouse model and a genetically engineered mouse model (GEMM). Activation of immune responses after administration of Kras vaccine and avasimibe was also assessed by flow cytometry, ELISpot and IHC. Findings We found that Kras vaccine combined with avasimibe significantly decreased the presence of regulatory T cells in the tumor microenvironment and facilitated CD8+ T cell infiltration in tumor sites. Avasimibe also enhanced the efficacy of Kras vaccines target mutant Kras. Whereas the Kras vaccine significantly increased antigen-specific intracellular IFN-γ and granzyme B levels in CD8+ T cells, avasimibe significantly increased the number of tumor-infiltrating CD8+ T cells. Additionally, modulation of cholesterol metabolism was found to specifically impact in T cells, and not in cancer cells. Interpretation Avasimibe complements the efficacy of a multi-peptide Kras vaccine in controlling lung cancer development and growth. This treatment regimen represents a novel immunoprevention approach to prevent lung cancer.
Collapse
Affiliation(s)
- Jing Pan
- Center for Disease Prevention Research and Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Qi Zhang
- Center for Disease Prevention Research and Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Katie Palen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Li Wang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Lifen Qiao
- Center for Disease Prevention Research and Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bryon Johnson
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shizuko Sei
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Ronald A Lubet
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Yian Wang
- Center for Disease Prevention Research and Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ming You
- Center for Disease Prevention Research and Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
19
|
da Cunha BR, Domingos C, Stefanini ACB, Henrique T, Polachini GM, Castelo-Branco P, Tajara EH. Cellular Interactions in the Tumor Microenvironment: The Role of Secretome. J Cancer 2019; 10:4574-4587. [PMID: 31528221 PMCID: PMC6746126 DOI: 10.7150/jca.21780] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/25/2019] [Indexed: 02/06/2023] Open
Abstract
Over the past years, it has become evident that cancer initiation and progression depends on several components of the tumor microenvironment, including inflammatory and immune cells, fibroblasts, endothelial cells, adipocytes, and extracellular matrix. These components of the tumor microenvironment and the neoplastic cells interact with each other providing pro and antitumor signals. The tumor-stroma communication occurs directly between cells or via a variety of molecules secreted, such as growth factors, cytokines, chemokines and microRNAs. This secretome, which derives not only from tumor cells but also from cancer-associated stromal cells, is an important source of key regulators of the tumorigenic process. Their screening and characterization could provide useful biomarkers to improve cancer diagnosis, prognosis, and monitoring of treatment responses.
Collapse
Affiliation(s)
- Bianca Rodrigues da Cunha
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil
| | - Célia Domingos
- Department of Biomedical Sciences and Medicine, University of Algarve, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
| | - Ana Carolina Buzzo Stefanini
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil
| | - Tiago Henrique
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
| | - Giovana Mussi Polachini
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
| | - Pedro Castelo-Branco
- Department of Biomedical Sciences and Medicine, University of Algarve, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
- Algarve Biomedical Center, Gambelas, Faro, Portugal
| | - Eloiza Helena Tajara
- Department of Molecular Biology, School of Medicine of São José do Rio Preto/FAMERP, São José do Rio Preto, SP, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil
| |
Collapse
|
20
|
Rothbauer M, Charwat V, Bachmann B, Sticker D, Novak R, Wanzenböck H, Mathies RA, Ertl P. Monitoring transient cell-to-cell interactions in a multi-layered and multi-functional allergy-on-a-chip system. LAB ON A CHIP 2019; 19:1916-1921. [PMID: 31070645 DOI: 10.1039/c9lc00108e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We have developed a highly integrated lab-on-a-chip containing embedded electrical microsensors, μdegassers and pneumatically-actuated micropumps to monitor allergic hypersensitivity. Rapid antigen-mediated histamine release (e.g. s to min) and resulting muscle contraction (<30 min) is detected by connecting an immune compartment containing sensitized basophile cells to a vascular co-culture model.
Collapse
Affiliation(s)
- Mario Rothbauer
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria. and Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Verena Charwat
- Department of Biotechnology, University of Agricultural Resources and Life Sciences, Muthgasse 18, 1090 Vienna, Austria
| | - Barbara Bachmann
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria. and Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria and AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, 1200 Vienna, Austria
| | - Drago Sticker
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Richard Novak
- Department of Chemistry, University of California at Berkeley, Lewis Hall, Berkeley, California, USA
| | - Heinz Wanzenböck
- Faculty of Electrical Engineering, Vienna University of Technology, Gußhausstr. 25-25a, 1040 Vienna, Austria
| | - Richard A Mathies
- Department of Chemistry, University of California at Berkeley, Lewis Hall, Berkeley, California, USA
| | - Peter Ertl
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria. and Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
21
|
Sacca PA, Mazza ON, Scorticati C, Vitagliano G, Casas G, Calvo JC. Human Periprostatic Adipose Tissue: Secretome from Patients With Prostate Cancer or Benign Prostate Hyperplasia. Cancer Genomics Proteomics 2019; 16:29-58. [PMID: 30587498 DOI: 10.21873/cgp.20110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND/AIM Periprostatic adipose tissue (PPAT) directs tumour behaviour. Microenvironment secretome provides information related to its biology. This study was performed to identify secreted proteins by PPAT, from both prostate cancer and benign prostate hyperplasia (BPH) patients. PATIENTS AND METHODS Liquid chromatography-mass spectrometry-based proteomic analysis was performed in PPAT-conditioned media (CM) from patients with prostate cancer (CMs-T) (stage T3: CM-T3, stage T2: CM-T2) or benign disease (CM-BPH). RESULTS The highest number and diversity of proteins was identified in CM-T3. Locomotion was the biological process mainly associated to CMs-T and reproduction to CM-T3. Immune responses were enriched in CMs-T. Extracellular matrix and structural proteins were associated to CMs-T. CM-T3 was enriched in proteins with catalytic activity and CM-T2 in proteins with defense/immunity activity. Metabolism and energy pathways were enriched in CM-T3 and those with immune system functions in CMs-T. Transport proteins were enriched in CM-T2 and CM-BPH. CONCLUSION Proteins and pathways reported in this study could be useful to distinguish stages of disease and may become targets for novel therapies.
Collapse
Affiliation(s)
- Paula Alejandra Sacca
- Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina
| | - Osvaldo Néstor Mazza
- Department of Urology, School of Medicine, University of Buenos Aires, Clínical Hospital "José de San Martín", Buenos Aires, Argentina
| | - Carlos Scorticati
- Department of Urology, School of Medicine, University of Buenos Aires, Clínical Hospital "José de San Martín", Buenos Aires, Argentina
| | | | - Gabriel Casas
- Department of Pathology, Deutsches Hospital, Buenos Aires, Argentina
| | - Juan Carlos Calvo
- Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina.,Department of Biological Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Wang X, He Y, Ye Y, Zhao X, Deng S, He G, Zhu H, Xu N, Liang S. SILAC-based quantitative MS approach for real-time recording protein-mediated cell-cell interactions. Sci Rep 2018; 8:8441. [PMID: 29855483 PMCID: PMC5981645 DOI: 10.1038/s41598-018-26262-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/04/2018] [Indexed: 02/05/2023] Open
Abstract
In tumor microenvironment, interactions among multiple cell types are critical for cancer progression. To understand the molecular mechanisms of these complex interplays, the secreted protein analysis between malignant cancer cells and the surrounding nonmalignant stroma is a good viewpoint to investigate cell-cell interactions. Here, we developed two stable isotope labeling of amino acids in cell culture (SILAC)-based mass spectrometry (MS)/MS approaches termed spike-in SILAC and triple-SILAC to quantify changes of protein secretion level in a cell co-cultured system. Within the co-culture system of CT26 and Ana-1 cells, the spike-in SILAC and triple-SILAC MS approaches are sensitive to quantitatively measure protein secretion changes. Three representative quantified proteins (Galectin-1, Cathepsin L1 and Thrombospondin-1) by two SILAC-based MS methods were further validated by Western blotting, and the coming result matched well with SILACs’. We further applied these two SILACs to human cell lines, NCM460 and HT29 co-culture system, for evaluating the feasibility, which confirmed the spike-in and triple SILAC were capable of monitoring the changed secreted proteins of human cell lines. Considering these two strategies in time consuming, sample complexity and proteome coverage, the triple-SILAC way shows more efficiency and economy for real-time recording secreted protein levels in tumor microenvironment.
Collapse
Affiliation(s)
- Xixi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China.,Chengdu Center for Disease Control and Prevention, Chengdu, 610041, P. R. China
| | - Yu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Yang Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xinyu Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Shi Deng
- Department of Urinary Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, P. R. China
| | - Gu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, P. R. China
| | - Ningzhi Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China.,Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, P. R. China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China.
| |
Collapse
|
23
|
Liu X, Hu J, Li Y, Cao W, Wang Y, Ma Z, Li F. Mesenchymal stem cells expressing interleukin-18 inhibit breast cancer in a mouse model. Oncol Lett 2018; 15:6265-6274. [PMID: 29725393 PMCID: PMC5920279 DOI: 10.3892/ol.2018.8166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 01/31/2017] [Indexed: 12/14/2022] Open
Abstract
Development of an improved breast cancer therapy has been an elusive goal of cancer gene therapy for a long period of time. Human mesenchymal stem cells derived from umbilical cord (hUMSCs) genetically modified with the interleukin (IL)-18 gene (hUMSCs/IL-18) were previously demonstrated to be able to suppress the proliferation, migration and invasion of breast cancer cells in vitro. In the present study, the effect of hUMSCs/IL-18 on breast cancer in a mouse model was investigated. A total of 128 mice were divided into 2 studies (the early-effect study and the late-effect study), with 4 groups in each, including the PBS-, hUMSC-, hUMSC/vector- and hUMSC/IL-18-treated groups. All treatments were injected along with 200 µl PBS. Following therapy, the tumor size, histological examination, and expression of lymphocytes, Ki-67, cluster of differentiation 31 and cytokines [interleukin (IL)-18, IL-12, interferon (IFN)-γ and TNF-α] in each group were analyzed. Proliferation of cells (assessed by measuring tumor size and Ki-67 expression) and metastasis, (by determining pulmonary and hepatic metastasis) of breast cancer cells in the hUMSC/IL-18 group were significantly decreased compared with all other groups. hUMSCs/IL-18 suppressed tumor cell proliferation by activating immunocytes and immune cytokines, decreasing the proliferation index of proliferation marker protein Ki-67 of tumor cells and inhibiting tumor angiogenesis. Furthermore, hUMSCs/IL-18 were able to induce a more marked and improved therapeutic effect in the tumor sites, particularly in early tumors. The results of the present study indicate that hUMSCs/IL-18 were able to inhibit the proliferation and metastasis of breast cancer cells in vivo, possibly leading to an approach for a novel antitumor therapy in breast cancer.
Collapse
Affiliation(s)
- Xiaoyi Liu
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jianxia Hu
- Stem Cell Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yueyun Li
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Weihong Cao
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yu Wang
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Zhongliang Ma
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Funian Li
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
24
|
Luo D, Hu S, Tang C, Liu G. Mesenchymal stem cells promote cell invasion and migration and autophagy-induced epithelial-mesenchymal transition in A549 lung adenocarcinoma cells. Cell Biochem Funct 2018; 36:88-94. [PMID: 29372557 DOI: 10.1002/cbf.3320] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/16/2017] [Accepted: 12/26/2017] [Indexed: 01/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are recruited into the tumour microenvironment and promote tumour growth and metastasis. Tumour microenvironment-induced autophagy is considered to suppress primary tumour formation by impairing migration and invasion. Whether these recruited MSCs regulate tumour autophagy and whether autophagy affects tumour growth are controversial. Our data showed that MSCs promote autophagy activation, reactive oxygen species production, and epithelial-mesenchymal transition (EMT) as well as increased migration and invasion in A549 cells. Decreased expression of E-cadherin and increased expression of vimentin and Snail were observed in A549 cells cocultured with MSCs. Conversely, MSC coculture-mediated autophagy positively promoted tumour EMT. Autophagy inhibition suppressed MSC coculture-mediated EMT and reduced A549 cell migration and invasion slightly. Furthermore, the migratory and invasive abilities of A549 cells were additional increased when autophagy was further enhanced by rapamycin treatment. Taken together, this work suggests that microenvironments containing MSCs can promote autophagy activation for enhancing EMT; MSCs also increase the migratory and invasive abilities of A549 lung adenocarcinoma cells. Mesenchymal stem cell-containing microenvironments and MSC-induced autophagy signalling may be potential targets for blocking lung cancer cell migration and invasion.
Collapse
Affiliation(s)
- Dan Luo
- Quality Control Section, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Shiyuan Hu
- Department of Orthopedics, The Third Affiliated Hospital, Army Medical University, Chongqing, China
| | - Chunlan Tang
- Quality Control Section, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guoxiang Liu
- Department of Respiration, Army Medical University, Chongqing, China
| |
Collapse
|
25
|
Prina-Mello A, Jain N, Liu B, Kilpatrick JI, Tutty MA, Bell AP, Jarvis SP, Volkov Y, Movia D. Culturing substrates influence the morphological, mechanical and biochemical features of lung adenocarcinoma cells cultured in 2D or 3D. Tissue Cell 2017; 50:15-30. [PMID: 29429514 DOI: 10.1016/j.tice.2017.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/31/2017] [Accepted: 11/26/2017] [Indexed: 01/04/2023]
Abstract
Alternative models such as three-dimensional (3D) cell cultures represent a distinct milestone towards capturing the realities of cancer biology in vitro and reduce animal experimentation in the preclinical stage of drug discovery. Significant work remains to be done to understand how substrates used in in vitro alternatives influence cancer cells phenotype and drug efficacy responses, so that to accurately link such models to specific in vivo disease scenarios. Our study describes how the morphological, mechanical and biochemical properties of adenocarcinoma (A549) cells change in response to a 3D environment and varying substrates. Confocal Laser Scanning (LSCM), He-Ion (HIM) and Atomic Force (AFM) microscopies, supported by ELISA and Western blotting, were used. These techniques enabled us to evaluate the shape, cytoskeletal organization, roughness, stiffness and biochemical signatures of cells grown within soft 3D matrices (PuraMatrix™ and Matrigel™), and to compare them to those of cells cultured on two-dimensional glass substrates. Cell cultures are also characterized for their biological response to docetaxel, a taxane-type drug used in Non-Small-Cell Lung Cancer (NSCLC) treatment. Our results offer an advanced biophysical insight into the properties and potential application of 3D cultures of A549 cells as in vitro alternatives in lung cancer research.
Collapse
Affiliation(s)
- Adriele Prina-Mello
- CRANN Institute and AMBER Centre, Trinity College Dublin, Ireland; Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Ireland; Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
| | - Namrata Jain
- CRANN Institute and AMBER Centre, Trinity College Dublin, Ireland
| | - Baiyun Liu
- School of Physics, University College Dublin, Ireland
| | - Jason I Kilpatrick
- Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Ireland
| | - Melissa A Tutty
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
| | - Alan P Bell
- CRANN Institute and AMBER Centre, Trinity College Dublin, Ireland; Advanced Microscopy Laboratory (AML), Trinity College Dublin, Ireland
| | - Suzanne P Jarvis
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland; School of Physics, University College Dublin, Ireland
| | - Yuri Volkov
- CRANN Institute and AMBER Centre, Trinity College Dublin, Ireland; Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Ireland; Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
| | - Dania Movia
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Ireland; Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland.
| |
Collapse
|
26
|
Chen Y, Zhang X. Pivotal regulators of tissue homeostasis and cancer: macrophages. Exp Hematol Oncol 2017; 6:23. [PMID: 28804688 PMCID: PMC5549331 DOI: 10.1186/s40164-017-0083-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022] Open
Abstract
Macrophages are an essential component of innate immunity and play a vital role in inflammation and host defense. Based on immunological responses, the macrophages are classified into "activated" macrophage (M1 macrophages) participating in the responses of type I helper T (Th1) cells to pathogens and "alternatively activated" macrophages (M2 macrophages) in response to interleukin (IL)-4 and IL-13. In this review, we discuss the origin, classification and function of macrophages. We also discuss the mechanisms underlying polarization of different macrophage subtypes, including transcriptional, epigenetic and post-transcriptional regulation.
Collapse
Affiliation(s)
- Yulei Chen
- College of Life Sciences, Zhejiang University, Hangzhou, 310058 People's Republic of China
| | - Xiaobo Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, 310058 People's Republic of China
| |
Collapse
|
27
|
Chen Y, Zhang S, Wang Q, Zhang X. Tumor-recruited M2 macrophages promote gastric and breast cancer metastasis via M2 macrophage-secreted CHI3L1 protein. J Hematol Oncol 2017; 10:36. [PMID: 28143526 PMCID: PMC5286803 DOI: 10.1186/s13045-017-0408-0] [Citation(s) in RCA: 300] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 01/25/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The macrophage, one of the several key immune cell types, is believed to be involved in tumorigenesis. However, the mechanism of macrophages promoting tumor progression is largely unknown. METHODS The differentially secreted proteins of M1 and M2 macrophages were analyzed by mass spectrometry. We performed GST pull-down assay for the identification of cell-membrane receptors that interact with chitinase 3-like protein 1 (CHI3L1) protein. The mouse model was used to validate the function of CHI3L1 in cancer metastasis in vivo. Protein phosphorylation and gene expression were performed to study the signaling pathway activation of cancer cells after CHI3L1 treatment. RESULTS M2 macrophage-secreted CHI3L1 promoted the metastasis of gastric and breast cancer cells in vitro and in vivo. The CHI3L1 protein functioned by interacting with interleukin-13 receptor α2 chain (IL-13Rα2) molecules on the plasma membranes of cancer cells. Activation of IL-13Rα2 by CHI3L1 triggered the activation of the mitogen-activated protein kinase signaling pathway, leading to the upregulated expression of matrix metalloproteinase genes, which promoted tumor metastasis. The results of this study indicated that the level of CHI3L1 protein in the sera of patients with gastric or breast cancer was significantly elevated compared with those of healthy donors. CONCLUSIONS Our study revealed a novel aspect of macrophages with respect to cancer metastasis and showed that CHI3L1 could be a marker of metastatic gastric and breast cancer in patients.
Collapse
Affiliation(s)
- Yulei Chen
- College of Life Sciences and Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Siyuan Zhang
- College of Life Sciences and Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Qizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233030, People's Republic of China
| | - Xiaobo Zhang
- College of Life Sciences and Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
28
|
Richards CD. Innate Immune Cytokines, Fibroblast Phenotypes, and Regulation of Extracellular Matrix in Lung. J Interferon Cytokine Res 2017; 37:52-61. [PMID: 28117653 DOI: 10.1089/jir.2016.0112] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation can be caused by adaptive immune responses in autoimmune and allergic conditions, driven by a T lymphocyte subset balance (TH1, TH2, Th17, Th22, and/or Treg) and skewed cellular profiles in an antigen-specific manner. However, several chronic inflammatory diseases have no clearly defined adaptive immune mechanisms that drive chronicity. These conditions include those that affect the lung such as nonatopic asthma or idiopathic pulmonary fibrosis comprising significant health problems. The remodeling of extracellular matrix (ECM) causes organ dysfunction, and it is largely generated by fibroblasts as the major cell controlling net ECM. As such, these are potential targets of treatment approaches in the context of ECM pathology. Fibroblast phenotypes contribute to ECM and inflammatory cell accumulation, and they are integrated into chronic disease mechanisms including cancer. Evidence suggests that innate cytokine responses may be critical in nonallergic/nonautoimmune disease, and they enable environmental agent exposure mechanisms that are independent of adaptive immunity. Innate immune cytokines derived from macrophage subsets (M1/M2) and innate lymphoid cell (ILC) subsets can directly regulate fibroblast function. We also suggest that STAT3-activating gp130 cytokines can sensitize fibroblasts to the innate cytokine milieu to drive phenotypes and exacerbate existing adaptive responses. Here, we review evidence exploring innate cytokine regulation of fibroblast behavior.
Collapse
Affiliation(s)
- Carl D Richards
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, Canada
| |
Collapse
|
29
|
Yao Y, Zhou Y, Su X, Dai L, Yu L, Deng H, Gou L, Yang J. Establishment and characterization of intraperitoneal xenograft models by co-injection of human tumor cells and extracellular matrix gel. Oncol Lett 2015; 10:3450-3456. [PMID: 26788149 PMCID: PMC4665880 DOI: 10.3892/ol.2015.3764] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 07/21/2015] [Indexed: 02/05/2023] Open
Abstract
Establishing a feasible intraperitoneal (i.p.) xenograft model in nude mice is a good strategy to evaluate the antitumor effect of drugs in vivo. However, the manipulation of human cancer cells in establishing a stable peritoneal carcinomatosis model in nude mice is problematic. In the present study, the ovarian and colorectal peritoneal tumor models were successfully established in nude mice by co-injection of human tumor cells and extracellular matrix gel. In ovarian tumor models, the mean number tumor nodes was significantly higher in the experimental group (intraperitoneal tumor cell co-injection with ECM gel) compared with the PBS control group on the 30th day (21.0±3.0 vs. 3.6±2.5; P<0.05). The same results were observed in the colorectal peritoneal tumor models on the 28th day. The colorectal peritoneal tumor model was further used to evaluate the chemotherapy effect of irinotecan (CPT-11). The mean weight of peritoneal tumor nodes in CPT-11 treatment group was significantly less than that of the control group (0.81±0.16 vs. 2.18±0.21 g; P<0.05). The results confirmed the value of these i.p. xenograft models in nude mice as efficient and feasible tools for preclinical evaluation.
Collapse
Affiliation(s)
- Yuqin Yao
- Research Centre for Public Health and Preventive Medicine, West China School of Public Health, No. 4 West China Teaching Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China; Guangdong Zhongsheng Pharmaceutical Co., Ltd., Dongguan, Guangdong 523000, P.R. China
| | - Yongjun Zhou
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lin Yu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lantu Gou
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jinliang Yang
- Research Centre for Public Health and Preventive Medicine, West China School of Public Health, No. 4 West China Teaching Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
30
|
Berglund E, Daré E, Branca RM, Akcakaya P, Fröbom R, Berggren PO, Lui WO, Larsson C, Zedenius J, Orre L, Lehtiö J, Kim J, Bränström R. Secretome protein signature of human gastrointestinal stromal tumor cells. Exp Cell Res 2015; 336:158-70. [DOI: 10.1016/j.yexcr.2015.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 01/03/2023]
|
31
|
Rudisch A, Dewhurst MR, Horga LG, Kramer N, Harrer N, Dong M, van der Kuip H, Wernitznig A, Bernthaler A, Dolznig H, Sommergruber W. High EMT Signature Score of Invasive Non-Small Cell Lung Cancer (NSCLC) Cells Correlates with NFκB Driven Colony-Stimulating Factor 2 (CSF2/GM-CSF) Secretion by Neighboring Stromal Fibroblasts. PLoS One 2015; 10:e0124283. [PMID: 25919140 PMCID: PMC4412534 DOI: 10.1371/journal.pone.0124283] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/12/2015] [Indexed: 12/23/2022] Open
Abstract
We established co-cultures of invasive or non-invasive NSCLC cell lines and various types of fibroblasts (FBs) to more precisely characterize the molecular mechanism of tumor-stroma crosstalk in lung cancer. The HGF-MET-ERK1/2-CREB-axis was shown to contribute to the onset of the invasive phenotype of Calu-1 with HGF being secreted by FBs. Differential expression analysis of the respective mono- and co-cultures revealed an upregulation of NFκB-related genes exclusively in co-cultures with Calu-1. Cytokine Array- and ELISA-based characterization of the “cytokine fingerprints” identified CSF2 (GM-CSF), CXCL1, CXCL6, VEGF, IL6, RANTES and IL8 as being specifically upregulated in various co-cultures. Whilst CXCL6 exhibited a strictly FB-type-specific induction profile regardless of the invasiveness of the tumor cell line, CSF2 was only induced in co-cultures of invasive cell lines regardless of the partnered FB type. These cultures revealed a clear link between the induction of CSF2 and the EMT signature of the cancer cell line. The canonical NFκB signaling in FBs, but not in tumor cells, was shown to be responsible for the induced and constitutive CSF2 expression. In addition to CSF2, cytokine IL6, IL8 and IL1B, and chemokine CXCL1 and CXCL6 transcripts were also shown to be increased in co-cultured FBs. In contrast, their induction was not strictly dependent on the invasiveness of the co-cultured tumor cell. In a multi-reporter assay, additional signaling pathways (AP-1, HIF1-α, KLF4, SP-1 and ELK-1) were found to be induced in FBs co-cultured with Calu-1. Most importantly, no difference was observed in the level of inducibility of these six signaling pathways with regard to the type of FBs used. Finally, upon tumor fibroblast interaction the massive induction of chemokines such as CXCL1 and CXCL6 in FBs might be responsible for increased recruitment of a monocytic cell line (THP-1) in a transwell assay.
Collapse
MESH Headings
- Animals
- Cadherins/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Communication/drug effects
- Cell Line, Tumor
- Cytokines/metabolism
- Dermis/pathology
- Epithelial-Mesenchymal Transition/drug effects
- Epithelial-Mesenchymal Transition/genetics
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks/drug effects
- Genes, Reporter
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Hepatocyte Growth Factor/pharmacology
- Humans
- Inflammation/pathology
- Kruppel-Like Factor 4
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- NF-kappa B/metabolism
- Neoplasm Invasiveness
- Phenotype
- Proto-Oncogene Proteins c-met/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Stromal Cells/metabolism
- Up-Regulation/drug effects
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Albin Rudisch
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- Department of Microbiology, Immunobiology and Genetics, Center of Molecular Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | | | | | - Nina Kramer
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Nathalie Harrer
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Meng Dong
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Baden-Württemberg, Germany
| | - Heiko van der Kuip
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Baden-Württemberg, Germany
| | - Andreas Wernitznig
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Andreas Bernthaler
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Sommergruber
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- * E-mail:
| |
Collapse
|
32
|
Méndez O, Villanueva J. Challenges and opportunities for cell line secretomes in cancer proteomics. Proteomics Clin Appl 2015; 9:348-57. [PMID: 25418557 DOI: 10.1002/prca.201400131] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/06/2014] [Accepted: 11/19/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Olga Méndez
- Vall d'Hebron Institute of Oncology (VHIO); Barcelona Spain
| | | |
Collapse
|
33
|
Singla AK, Downey CM, Bebb GD, Jirik FR. Characterization of a murine model of metastatic human non-small cell lung cancer and effect of CXCR4 inhibition on the growth of metastases. Oncoscience 2015; 2:263-71. [PMID: 25897429 PMCID: PMC4394132 DOI: 10.18632/oncoscience.117] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/06/2015] [Indexed: 01/23/2023] Open
Abstract
Despite successful preclinical testing carried out through the use of subcutaneous xenografted tumors, many anti-cancer agents have gone on to fail in human trials. One potential factor accounting for this discrepancy may relate to the inadequacy of the commonly employed preclinical models to recapitulate the human disease, particularly when it comes to discovery of agents that are effective against advanced disease. Herein, we report the characterization of a NSCLC model and an exploration of the impact that a CXCR4 inhibitor, AMD3100, had on NCI-H1299-derived metastasis. These cells express a variety of metastasis-promoting factors, hence we selected them for a study of their metastatic colonization potential. To accomplish this, luciferase-expressing H1299 (H1299-luc2) cells were inoculated into athymic mice via the intracardiac route. This strategy produced adrenal, bone, ovarian, and pancreatic metastases, sites commonly involved in human metastatic NSCLC. Notably, micro-computed tomography and histological evaluation of the skeletal lesions revealed the presence of extensive osteolysis. To investigate the potential role of CXCR4 in mediating metastatic colonization of tissues, AMD3100 was administered to mice inoculated with H1299-luc2 cells. While this treatment did not appreciably alter the frequency of metastatic colonization, it was able to slow the growth of macrometastases. This model, recapitulating some of the events seen in late-stage human NSCLC, may prove useful in the evaluation of new therapies targeting metastatic disease.
Collapse
Affiliation(s)
- Arvind K Singla
- Department of Biochemistry and Molecular Biology, The McCaig Institute for Bone and Joint Health, Alberta, Canada
| | - Charlene M Downey
- Department of Biochemistry and Molecular Biology, The McCaig Institute for Bone and Joint Health, Alberta, Canada
| | - Gwyn D Bebb
- Tom Baker Cancer Centre, and Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Frank R Jirik
- Department of Biochemistry and Molecular Biology, The McCaig Institute for Bone and Joint Health, Alberta, Canada
| |
Collapse
|
34
|
A novel immunocompetent murine model for replicating oncolytic adenoviral therapy. Cancer Gene Ther 2014; 22:17-22. [PMID: 25525035 PMCID: PMC4298785 DOI: 10.1038/cgt.2014.64] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 01/23/2023]
Abstract
Oncolytic adenoviruses are under investigation as a promising novel strategy for cancer immunotherapeutics. Unfortunately, there is no immunocompetent mouse cancer model to test oncolytic adenovirus because murine cancer cells are generally unable to produce infectious viral progeny from human adenoviruses. We find that the murine K-ras-induced lung adenocarcinoma cell line ADS-12 supports adenoviral infection and generates infectious viral progeny. ADS-12 cells express the coxsackie and adenovirus receptor and infected ADS-12 cells express the viral protein E1A. We find that our previously described oncolytic virus, adenovirus TAV-255 (AdTAV-255), kills ADS-12 cells in a dose- and time-dependent manner. We investigated ADS-12 cells as an in-vivo model system for replicating oncolytic adenoviruses. Subcutaneous injection of ADS-12 cells into immunocompetent 129 mice led to tumor formation in all injected mice. Intratumoral injection of AdTAV-255 in established tumors causes a significant reduction in tumor growth. This model system represents the first fully immunocompetent mouse model for cancer treatment with replicating oncolytic adenoviruses, and therefore will be useful to study the therapeutic effect of oncolytic adenoviruses in general and particularly immunostimulatory viruses designed to evoke an antitumor immune response.
Collapse
|
35
|
Fritz JM, Tennis MA, Orlicky DJ, Lin H, Ju C, Redente EF, Choo KS, Staab TA, Bouchard RJ, Merrick DT, Malkinson AM, Dwyer-Nield LD. Depletion of tumor-associated macrophages slows the growth of chemically induced mouse lung adenocarcinomas. Front Immunol 2014; 5:587. [PMID: 25505466 PMCID: PMC4243558 DOI: 10.3389/fimmu.2014.00587] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 11/03/2014] [Indexed: 11/13/2022] Open
Abstract
Chronic inflammation is a risk factor for lung cancer, and low-dose aspirin intake reduces lung cancer risk. However, the roles that specific inflammatory cells and their products play in lung carcinogenesis have yet to be fully elucidated. In mice, alveolar macrophage numbers increase as lung tumors progress, and pulmonary macrophage programing changes within 2 weeks of carcinogen exposure. To examine how macrophages specifically affect lung tumor progression, they were depleted in mice bearing urethane-induced lung tumors using clodronate-encapsulated liposomes. Alveolar macrophage populations decreased to ≤50% of control levels after 4–6 weeks of liposomal clodronate treatment. Tumor burden decreased by 50% compared to vehicle treated mice, and tumor cell proliferation, as measured by Ki67 staining, was also attenuated. Pulmonary fluid levels of insulin-like growth factor-I, CXCL1, IL-6, and CCL2 diminished with clodronate liposome treatment. Tumor-associated macrophages expressed markers of both M1 and M2 programing in vehicle and clodronate liposome-treated mice. Mice lacking CCR2 (the receptor for macrophage chemotactic factor CCL2) had comparable numbers of alveolar macrophages and showed no difference in tumor growth rates when compared to similarly treated wild-type mice suggesting that while CCL2 may recruit macrophages to lung tumor microenvironments, redundant pathways can compensate when CCL2/CCR2 signaling is inactivated. Depletion of pulmonary macrophages rather than inhibition of their recruitment may be an advantageous strategy for attenuating lung cancer progression.
Collapse
Affiliation(s)
- Jason M Fritz
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver , Aurora, CO , USA
| | - Meredith A Tennis
- Pulmonary Division, School of Medicine, University of Colorado Denver , Aurora, CO , USA
| | - David J Orlicky
- Department of Pathology, School of Medicine, University of Colorado Denver , Aurora, CO , USA
| | - Hao Lin
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver , Aurora, CO , USA
| | - Cynthia Ju
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver , Aurora, CO , USA
| | | | - Kevin S Choo
- Research Division, Eastern Colorado Veterans Administration Medical Center , Denver, CO , USA
| | - Taylor A Staab
- Research Division, Eastern Colorado Veterans Administration Medical Center , Denver, CO , USA
| | - Ronald J Bouchard
- Research Division, Eastern Colorado Veterans Administration Medical Center , Denver, CO , USA
| | - Daniel T Merrick
- Department of Pathology, School of Medicine, University of Colorado Denver , Aurora, CO , USA
| | - Alvin M Malkinson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver , Aurora, CO , USA
| | - Lori D Dwyer-Nield
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver , Aurora, CO , USA
| |
Collapse
|
36
|
Zheng DJ, Yu GH, Gao JF, Gu JD. Concomitant EGFR inhibitors combined with radiation for treatment of non-small cell lung carcinoma. Asian Pac J Cancer Prev 2014; 14:4485-94. [PMID: 24083690 DOI: 10.7314/apjcp.2013.14.8.4485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is considered to be one of the key driver genes in non-small cell lung cancer (NSCLC). Several clinical trials have shown great promise of EGFR tyrosine kinase inhibitors (TKIs) in the first-line treatment of NSCLC. Many advances have been made in the understanding of EGFR signal transduction network and the interaction between EGFR and tumor microenvironment in mediating cancer survival and development. The concomitant targeted therapy and radiation is a new strategy in the treatment of NSCLC. A number of preclinical studies have demonstrated synergistic anti-tumor activity in the combination of EGFR inhibitors and radiotherapy in vitro and in vivo. In the present review, we discuss the rationale of the combination of EGFR inhibitors and radiotherapy in the treatment of NSCLC.
Collapse
Affiliation(s)
- De-Jie Zheng
- Department of Clinical Oncology, Weifang People's Hospital, Weifang, China E-mail :
| | | | | | | |
Collapse
|
37
|
Hanash S, Schliekelman M. Proteomic profiling of the tumor microenvironment: recent insights and the search for biomarkers. Genome Med 2014; 6:12. [PMID: 24713112 PMCID: PMC3978437 DOI: 10.1186/gm529] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although gain of oncogene functions and loss of tumor suppressor functions are driving forces in tumor development, the tumor microenvironment, comprising the extracellular matrix, surrounding stroma, signaling molecules and infiltrating immune and other cell populations, is now also recognized as crucial to tumor development and metastasis. Many interactions at the tumor cell-environment interface occur at the protein level. Proteomic approaches are contributing to the definition of the protein constituents of the microenvironment and their sources, modifications, interactions and turnover, as well as providing information on how these features relate to tumor development and progression. Recently, proteomic studies have revealed how cancer cells modulate the microenvironment through their secreted proteins and how they can alter their protein constituents to adapt to the microenvironment. Moreover, the release of proteins from the microenvironment into the circulatory system has relevance for the development of blood-based cancer diagnostics. Here, we review how proteomic approaches are being applied to studies of the tumor microenvironment to decipher tumor-stroma interactions and to elucidate the role of host cells in the tumor microenvironment.
Collapse
|
38
|
Costa E, Faiad O, Landgraf R, Ferreira A, Brigatte P, Curi R, Cury Y, Sampaio S. Involvement of formyl peptide receptors in the stimulatory effect of crotoxin on macrophages co-cultivated with tumour cells. Toxicon 2013; 74:167-78. [DOI: 10.1016/j.toxicon.2013.08.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 06/27/2013] [Accepted: 08/13/2013] [Indexed: 12/01/2022]
|
39
|
Quantitative secretome analysis reveals the interactions between epithelia and tumor cells by in vitro modulating colon cancer microenvironment. J Proteomics 2013; 89:51-70. [DOI: 10.1016/j.jprot.2013.05.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/22/2013] [Accepted: 05/20/2013] [Indexed: 11/18/2022]
|
40
|
Expression of CXCL1 in human endothelial cells induces angiogenesis through the CXCR2 receptor and the ERK1/2 and EGF pathways. J Transl Med 2013; 93:768-78. [PMID: 23732813 DOI: 10.1038/labinvest.2013.71] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Endothelial cell growth and proliferation are critical for angiogenesis; thus, greater insight into the regulation of pathological angiogenesis is greatly needed. Previous studies have reported on chemokine (C-X-C motif) ligand 1 (CXCL1) expression in epithelial cells and that secretion of CXCL1 from these epithelial cells induces angiogenesis. However, limited reports have demonstrated CXCL1 expression in endothelial cells. In this report, we present data that expand on the role of CXCL1 in human endothelial cells inducing angiogenesis. Specifically, CXCL1 is expressed and secreted from human endothelial cells. Interference of CXCL1 function using neutralizing antibodies resulted in a reduction in endothelial cell migration and viability/proliferation, the latter associated with a decrease in levels of cyclin D and cdk4. In vitro studies revealed that CXCL1 influenced neoangiogenesis through the regulation of epidermal growth factor and ERK1/2. In a xenograft angiogenesis model, interference of CXCL1 function resulted in inhibition of angiogenesis. A better understanding of the role of CXCL1 in the interactions between the endothelial and epithelial components will provide insight into how human tissues use CXCL1 to survive and thrive in a hostile environment.
Collapse
|
41
|
Holmberg C, Ghesquière B, Impens F, Gevaert K, Kumar JD, Cash N, Kandola S, Hegyi P, Wang TC, Dockray GJ, Varro A. Mapping proteolytic processing in the secretome of gastric cancer-associated myofibroblasts reveals activation of MMP-1, MMP-2, and MMP-3. J Proteome Res 2013; 12:3413-22. [PMID: 23705892 PMCID: PMC3709265 DOI: 10.1021/pr400270q] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cancer progression involves changes in extracellular proteolysis, but the contribution of stromal cell secretomes to the cancer degradome remains uncertain. We have now defined the secretome of a specific stromal cell type, the myofibroblast, in gastric cancer and its modification by proteolysis. SILAC labeling and COFRADIC isolation of methionine containing peptides allowed us to quantify differences in gastric cancer-derived myofibroblasts compared with myofibroblasts from adjacent tissue, revealing increased abundance of several proteases in cancer myofibroblasts including matrix metalloproteinases (MMP)-1 and -3. Moreover, N-terminal COFRADIC analysis identified cancer-restricted proteolytic cleavages, including liberation of the active forms of MMP-1, -2, and -3 from their inactive precursors. In vivo imaging confirmed increased MMP activity when gastric cancer cells were xenografted in mice together with gastric cancer myofibroblasts. Western blot and enzyme activity assays confirmed increased MMP-1, -2, and -3 activity in cancer myofibroblasts, and cancer cell migration assays indicated stimulation by MMP-1, -2, and -3 in cancer-associated myofibroblast media. Thus, cancer-derived myofibroblasts differ from their normal counterparts by increased production and activation of MMP-1, -2, and -3, and this may contribute to the remodelling of the cancer cell microenvironment.
Collapse
|
42
|
Lo HM, Shieh JM, Chen CL, Tsou CJ, Wu WB. Vascular endothelial growth factor induces CXCL1 chemokine release via JNK and PI-3K-dependent pathways in human lung carcinoma epithelial cells. Int J Mol Sci 2013; 14:10090-106. [PMID: 23665907 PMCID: PMC3676830 DOI: 10.3390/ijms140510090] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 04/23/2013] [Accepted: 05/02/2013] [Indexed: 12/14/2022] Open
Abstract
Lung cancer cells express different chemokines and chemokine receptors that modulate leukocyte infiltration within tumor microenvironment. In this study we screened several mediators/growth factors on CXCL1 release in human carcinoma epithelial cells. Of the tested mediators, VEGF was found to have a robust increase in causing CXCL1 release. VEGF stimulated CXCL1 release and mRNA expression in a time- and concentration-dependent manner. The release was inhibited by the VEGF receptor antagonists and the JNK, PI-3K, tyrosine kinase, and transcription inhibitors. In parallel, VEGF induced JNK, PI3K and Akt activation. Strikingly, among these inhibitors only the JNK inhibitor could reduce VEGF-induced CXCL1 mRNA expression, suggesting that JNK participated in VEGF-induced CXCL1 synthesis, whereas PI-3K was responsible for cellular CXCL1 secretory process. In addition, the steroid dexamethasone and TGF-β suppressed CXCL1 release through a transcriptional regulation. We also showed that cells stimulated with VEGF significantly attracted monocyte migration, which could be abolished by CXCL1 B/N Ab, CXC receptor 2 antagonist, TGF-β, and dexamethasone. In summary, we provide here evidence showing JNK activation for VEGF-induced CXCL1 DNA transcription and PI-3K pathway for extracellular CXCL1 release in human carcinoma epithelial cells. The released CXCL1 was functionally linked to recruiting monocytes into lung cancer cell microenvironment.
Collapse
Affiliation(s)
- Huey-Ming Lo
- School of Medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan; E-Mails: (H.-M.L.); (C.-L.C.); (C.-J.T.)
- Section of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan
| | - Jiunn-Min Shieh
- Department of Internal Medicine, Chi-Mei Medical Center, Tainan 71004, Taiwan; E-Mail:
| | - Chih-Li Chen
- School of Medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan; E-Mails: (H.-M.L.); (C.-L.C.); (C.-J.T.)
| | - Chih-Jen Tsou
- School of Medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan; E-Mails: (H.-M.L.); (C.-L.C.); (C.-J.T.)
| | - Wen-Bin Wu
- School of Medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan; E-Mails: (H.-M.L.); (C.-L.C.); (C.-J.T.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-2-2905-3497; Fax: +886-2-2905-2096
| |
Collapse
|
43
|
Speisman RB, Kumar A, Rani A, Foster TC, Ormerod BK. Daily exercise improves memory, stimulates hippocampal neurogenesis and modulates immune and neuroimmune cytokines in aging rats. Brain Behav Immun 2013; 28:25-43. [PMID: 23078985 PMCID: PMC3545095 DOI: 10.1016/j.bbi.2012.09.013] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 09/19/2012] [Accepted: 09/26/2012] [Indexed: 01/02/2023] Open
Abstract
We tested whether daily exercise modulates immune and neuroimmune cytokines, hippocampus-dependent behavior and hippocampal neurogenesis in aging male F344 rats (18mo upon arrival). Twelve weeks after conditioned running or control group assignment, the rats were trained and tested in a rapid water maze followed by an inhibitory avoidance task. The rats were BrdU-injected beginning 12days after behavioral testing and killed 3weeks later to quantify cytokines and neurogenesis. Daily exercise increased neurogenesis and improved immediate and 24h water maze discrimination index (DI) scores and 24h inhibitory avoidance retention latencies. Daily exercise decreased cortical VEGF, hippocampal IL-1β and serum MCP-1, GRO-KC and leptin levels but increased hippocampal GRO-KC and IL-18 concentrations. Serum leptin concentration correlated negatively with new neuron number and both DI scores while hippocampal IL-1β concentration correlated negatively with memory scores in both tasks. Cortical VEGF, serum GRO-KC and serum MCP-1 levels correlated negatively with immediate DI score and we found novel positive correlations between hippocampal IL-18 and GRO-KC levels and new neuron number. Pathway analyses revealed distinct serum, hippocampal and cortical compartment cytokine relationships. Our results suggest that daily exercise potentially improves cognition in aging rats by modulating hippocampal neurogenesis and immune and neuroimmune cytokine signaling. Our correlational data begin to provide a framework for systematically manipulating these immune and neuroimmune signaling molecules to test their effects on cognition and neurogenesis across lifespan in future experiments.
Collapse
Affiliation(s)
- Rachel. B. Speisman
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Ashok Kumar
- Department of Neuroscience, University of Florida, Gainesville, FL, USA,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Asha Rani
- Department of Neuroscience, University of Florida, Gainesville, FL, USA,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Thomas C. Foster
- Department of Neuroscience, University of Florida, Gainesville, FL, USA,McKnight Brain Institute, University of Florida, Gainesville, FL, USA,Corresponding Author: Brandi K. Ormerod, PhD, Assistant Professor, J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-6131, USA, Phone: 352-273-8125, Fax: 352-273-9221,
| | - Brandi K. Ormerod
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA,Department of Neuroscience, University of Florida, Gainesville, FL, USA,McKnight Brain Institute, University of Florida, Gainesville, FL, USA,Corresponding Author: Brandi K. Ormerod, PhD, Assistant Professor, J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611-6131, USA, Phone: 352-273-8125, Fax: 352-273-9221,
| |
Collapse
|
44
|
Lin Q, Tan HT, Lim HSR, Chung MCM. Sieving through the cancer secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2360-71. [PMID: 23376431 DOI: 10.1016/j.bbapap.2013.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/03/2013] [Accepted: 01/24/2013] [Indexed: 12/22/2022]
Abstract
Cancer is among the most prevalent and serious health problems worldwide. Therefore, there is an urgent need for novel cancer biomarkers with high sensitivity and specificity for early detection and management of the disease. The cancer secretome, encompassing all the proteins that are secreted by cancer cells, is a promising source of biomarkers as the secreted proteins are most likely to enter the blood circulation. Moreover, since secreted proteins are responsible for signaling and communication with the tumor microenvironment, studying the cancer secretome would further the understanding of cancer biology. Latest developments in proteomics technologies have significantly advanced the study of the cancer secretome. In this review, we will present an overview of the secretome sample preparation process and summarize the data from recent secretome studies of six common cancers with high mortality (breast, colorectal, gastric, liver, lung and prostate cancers). In particular, we will focus on the various platforms that were employed and discuss the clinical applicability of the key findings in these studies. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Qifeng Lin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, 117597 Singapore
| | | | | | | |
Collapse
|
45
|
Schaaij-Visser TBM, de Wit M, Lam SW, Jiménez CR. The cancer secretome, current status and opportunities in the lung, breast and colorectal cancer context. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2242-58. [PMID: 23376433 DOI: 10.1016/j.bbapap.2013.01.029] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 12/20/2022]
Abstract
Despite major improvements on the knowledge and clinical management, cancer is still a deadly disease. Novel biomarkers for better cancer detection, diagnosis and treatment prediction are urgently needed. Proteins secreted, shed or leaking from the cancer cell, collectively termed the cancer secretome, are promising biomarkers since they might be detectable in blood or other biofluids. Furthermore, the cancer secretome in part represents the tumor microenvironment that plays a key role in tumor promoting processes such as angiogenesis and invasion. The cancer secretome, sampled as conditioned medium from cell lines, tumor/tissue interstitial fluid or tumor proximal body fluids, can be studied comprehensively by nanoLC-MS/MS-based approaches. Here, we outline the importance of current cancer secretome research and describe the mass spectrometry-based analysis of the secretome. Further, we provide an overview of cancer secretome research with a focus on the three most common cancer types: lung, breast and colorectal cancer. We conclude that the cancer secretome research field is a young, but rapidly evolving research field. Up to now, the focus has mainly been on the discovery of novel promising secreted cancer biomarker proteins. An interesting finding that merits attention is that in cancer unconventional secretion, e.g. via vesicles, seems increased. Refinement of current approaches and methods and progress in clinical validation of the current findings are vital in order to move towards applications in cancer management. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Tieneke B M Schaaij-Visser
- OncoProteomics Laboratory, Dept. of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands; Division of Molecular Genetics and Centre for Biomedical Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
46
|
Vicent S, Sayles LC, Vaka D, Khatri P, Gevaert O, Chen R, Zheng Y, Gillespie AK, Clarke N, Xu Y, Shrager J, Hoang CD, Plevritis S, Butte AJ, Sweet-Cordero EA. Cross-species functional analysis of cancer-associated fibroblasts identifies a critical role for CLCF1 and IL-6 in non-small cell lung cancer in vivo. Cancer Res 2012; 72:5744-56. [PMID: 22962265 DOI: 10.1158/0008-5472.can-12-1097] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cancer-associated fibroblasts (CAF) have been reported to support tumor progression by a variety of mechanisms. However, their role in the progression of non-small cell lung cancer (NSCLC) remains poorly defined. In addition, the extent to which specific proteins secreted by CAFs contribute directly to tumor growth is unclear. To study the role of CAFs in NSCLCs, a cross-species functional characterization of mouse and human lung CAFs was conducted. CAFs supported the growth of lung cancer cells in vivo by secretion of soluble factors that directly stimulate the growth of tumor cells. Gene expression analysis comparing normal mouse lung fibroblasts and mouse lung CAFs identified multiple genes that correlate with the CAF phenotype. A gene signature of secreted genes upregulated in CAFs was an independent marker of poor survival in patients with NSCLC. This secreted gene signature was upregulated in normal lung fibroblasts after long-term exposure to tumor cells, showing that lung fibroblasts are "educated" by tumor cells to acquire a CAF-like phenotype. Functional studies identified important roles for CLCF1-CNTFR and interleukin (IL)-6-IL-6R signaling in promoting growth of NSCLCs. This study identifies novel soluble factors contributing to the CAF protumorigenic phenotype in NSCLCs and suggests new avenues for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Silvestre Vicent
- AutCancer Biology Program, Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jin L, Zhang Y, Li H, Yao L, Fu D, Yao X, Xu LX, Hu X, Hu G. Differential secretome analysis reveals CST6 as a suppressor of breast cancer bone metastasis. Cell Res 2012; 22:1356-73. [PMID: 22688893 DOI: 10.1038/cr.2012.90] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bone metastasis is a frequent complication of breast cancer and a common cause of morbidity and mortality from the disease. During metastasis secreted proteins play crucial roles in the interactions between cancer cells and host stroma. To characterize the secreted proteins that are associated with breast cancer bone metastasis, we preformed a label-free proteomic analysis to compare the secretomes of four MDA-MB-231 (MDA231) derivative cell lines with varied capacities of bone metastasis. A total of 128 proteins were found to be consistently up-/down-regulated in the conditioned medium of bone-tropic cancer cells. The enriched molecular functions of the altered proteins included receptor binding and peptidase inhibition. Through additional transcriptomic analyses of breast cancer cells, we selected cystatin E/M (CST6), a cysteine protease inhibitor down-regulated in bone-metastatic cells, for further functional studies. Our results showed that CST6 suppressed the proliferation, colony formation, migration and invasion of breast cancer cells. The suppressive function against cancer cell motility was carried out by cancer cell-derived soluble CST6. More importantly, ectopic expression of CST6 in cancer cells rescued mice from overt osteolytic metastasis and deaths in the animal study, while CST6 knockdown markedly enhanced cancer cell bone metastasis and shortened animal survival. Overall, our study provided a systemic secretome analysis of breast cancer bone tropism and established secreted CST6 as a bona fide suppressor of breast cancer osteolytic metastasis.
Collapse
Affiliation(s)
- Lei Jin
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Rd, Shanghai 200025, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Fritz JM, Dwyer-Nield LD, Malkinson AM. Stimulation of neoplastic mouse lung cell proliferation by alveolar macrophage-derived, insulin-like growth factor-1 can be blocked by inhibiting MEK and PI3K activation. Mol Cancer 2011; 10:76. [PMID: 21699731 PMCID: PMC3135566 DOI: 10.1186/1476-4598-10-76] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 06/24/2011] [Indexed: 11/30/2022] Open
Abstract
Background Worldwide, lung cancer kills more people than breast, colon and prostate cancer combined. Alterations in macrophage number and function during lung tumorigenesis suggest that these immune effector cells stimulate lung cancer growth. Evidence from cancer models in other tissues suggests that cancer cells actively recruit growth factor-producing macrophages through a reciprocal signaling pathway. While the levels of lung macrophages increase during tumor progression in mouse models of lung cancer, and high pulmonary macrophage content correlates with a poor prognosis in human non-small cell lung cancer, the specific role of alveolar macrophages in lung tumorigenesis is not clear. Methods After culturing either an immortalized lung macrophage cell line or primary murine alveolar macrophages from naïve and lung-tumor bearing mice with primary tumor isolates and immortalized cell lines, the effects on epithelial proliferation and cellular kinase activation were determined. Insulin-like growth factor-1 (IGF-1) was quantified by ELISA, and macrophage conditioned media IGF-1 levels manipulated by IL-4 treatment, immuno-depletion and siRNA transfection. Results Primary macrophages from both naïve and lung-tumor bearing mice stimulated epithelial cell proliferation. The lungs of tumor-bearing mice contained 3.5-times more IGF-1 than naïve littermates, and media conditioned by freshly isolated tumor-educated macrophages contained more IGF-1 than media conditioned by naïve macrophages; IL-4 stimulated IGF-1 production by both macrophage subsets. The ability of macrophage conditioned media to stimulate neoplastic proliferation correlated with media IGF-1 levels, and recombinant IGF-1 alone was sufficient to induce epithelial proliferation in all cell lines evaluated. Macrophage-conditioned media and IGF-1 stimulated lung tumor cell growth in an additive manner, while EGF had no effect. Macrophage-derived factors increased p-Erk1/2, p-Akt and cyclin D1 levels in neoplastic cells, and the combined inhibition of both MEK and PI3K ablated macrophage-mediated increases in epithelial growth. Conclusions Macrophages produce IGF-1 which directly stimulates neoplastic proliferation through Erk and Akt activation. This observation suggests that combining macrophage ablation therapy with IGF-1R, MEK and/or PI3K inhibition could improve therapeutic response in human lung cancer. Exploring macrophage-based intervention could be a fruitful avenue for future research.
Collapse
Affiliation(s)
- Jason M Fritz
- Department of Pharmaceutical Sciences, Skagg School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E, Montview Blvd, C-238 V20-4460, Aurora, CO 80045, USA
| | | | | |
Collapse
|
49
|
VEGF and IL-18 in induced sputum of lung cancer patients. Cytokine 2011; 54:277-81. [DOI: 10.1016/j.cyto.2011.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2010] [Revised: 01/20/2011] [Accepted: 02/15/2011] [Indexed: 12/22/2022]
|
50
|
Fallica B, Makin G, Zaman MH. Bioengineering approaches to study multidrug resistance in tumor cells. Integr Biol (Camb) 2011; 3:529-39. [PMID: 21387035 DOI: 10.1039/c0ib00142b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The ability of cancer cells to become resistant to chemotherapeutic agents is a major challenge for the treatment of malignant tumors. Several strategies have emerged to attempt to inhibit chemoresistance, but the fact remains that resistance is a problem for every effective anticancer drug. The first part of this review will focus on the mechanisms of chemoresistance. It is important to understand the environmental cues, transport limitations and the cellular signaling pathways associated with chemoresistance before we can hope to effectively combat it. The second part of this review focuses on the work that needs to be done moving forward. Specifically, this section focuses on the necessity of translational research and interdisciplinary directives. It is critical that the expertise of oncologists, biologists, and engineers be brought together to attempt to tackle the problem. This discussion is from an engineering perspective, as the dialogue between engineers and other cancer researchers is the most challenging due to non-overlapping background knowledge. Chemoresistance is a complex and devastating process, meaning that we urgently need sophisticated methods to study the process of how cells become resistant.
Collapse
Affiliation(s)
- Brian Fallica
- Department of Biomedical Engineering, Boston University, USA
| | | | | |
Collapse
|