1
|
Miao C, Liu L, Cao Y, Jiang Z, Ding Z, Chen Y, Li H, Ma Z, Ma P, Zhang G, Li L, Li C. OSCC-derived EVs educate fibroblasts and remodel collagen landscape. Matrix Biol 2024:S0945-053X(24)00125-2. [PMID: 39393503 DOI: 10.1016/j.matbio.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/13/2024]
Abstract
Cancer-associated myofibroblasts (mCAFs) represent a significant component of the tumor microenvironment due to their contributions to extracellular matrix (ECM) remodeling. The pro-tumor mechanisms of extracellular vesicles (EVs) by regulating mCAFs and related collagens remain poorly understood in oral squamous cell carcinoma (OSCC). In this study, through analysis of single-cell sequencing data and immunofluorescence staining, we confirmed the increased presence of mCAFs and enrichment of specific collagen types in OSCC tissues. Furthermore, we demonstrated that OSCC-derived EVs promote the transformation of fibroblasts into mCAFs, leading to tumor invasion. Proteomic analysis identified the presence of TGF-β1 in EVs and revealed its role in inducing mCAFs via the TGF-β1/Smad3 signaling pathway. Experiments in vivo confirmed that EVs, particularly those carrying TGF-β1, trigger COL18high COL5high matrix deposition, thereby forming the pro-tumor ECM in OSCC. In summary, our investigation unveils the significant involvement of OSCC-derived EVs in orchestrating the differentiation of fibroblasts into mCAFs and modulating specific collagen types within the ECM. Therefore, this study provides a theoretical basis for targeting the EV-mediated TGF-β1 signaling pathway as a potential therapeutic strategy for OSCC.
Collapse
Affiliation(s)
- Cheng Miao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University
| | - Liu Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University
| | - Yubin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University.
| | - Zhishen Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University
| | - Zhangfan Ding
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University
| | - Yafei Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University
| | - Honglin Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University
| | - Zhongkai Ma
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University
| | - Pingchuan Ma
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University
| | - Gaowei Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University
| | - Longjiang Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University.
| | - Chunjie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University.
| |
Collapse
|
2
|
Malik S, Waquar S, Idrees N, Malik A. Impending role of inflammatory markers and their specificity and sensitivity in breast cancer patients. Sci Rep 2024; 14:15117. [PMID: 38956273 PMCID: PMC11219843 DOI: 10.1038/s41598-024-65821-8] [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: 12/17/2023] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Cancer and related disorders are the most common cause of cancer-related mortality with the incidence of 1 in 9 among the pre-menopausal Pakistani females. among the most common ailments worldwide, indicating the importance of developing particular techniques that could help attenuate the effects of breast cancer and related outcomes. The primary aim of the current study was to review the role of inflammatory and stress markers in the development and progression of breast cancer. Four hundred ninety-eight (n = 498) patients with breast cancer and four hundred and ninety-eight (n = 498) age- and sex-matched controls were selected for this case‒control study. Serum samples were obtained, and the levels of stress and inflammatory markers, including Matrix metalloproteases (MMPs), Interleukins (ILs), Heat shock proteins (HSPs), Malondialdehyde (MDA), Nitric Oxide (NO), inducible Nitric Oxide Synthase (iNOS) and Tumour necrosis factor-alpha (TNF-α), were determined. Most (62%) patients had metastatic breast cancer (stage III or IV) with an adverse grade (65% with Grade III and 35% with Grade II). The present study showed that the levels of oxidants such as MDA, ILs, MMPs and HSPs were significantly greater, while the levels of antioxidants such as Superoxide Dismutase (SOD), Glutathione (GSH), Catalase (CAT), vitamin A, C and D were significantly lower in breast cancer patients than in controls, suggesting their diagnostic importance and role in the pathophysiology of breast cancer. Oxidants, including IL-1, HSP27 and MMP9, which are highly specific and sensitive, may be used to develop the pathophysiological pathways of metastatic breast cancer in these patients. These pathways include cell invasion, cell migration and epithelial-mesenchymal transition. Therefore, we concluded that an increase in growth factors, e.g., Vascular Endothelial Growth Factor (VEGF), Tumour Growth Factor-beta (TGF-β) and B-cell lymphoma (Bcl2), under the influence of these variables plays a crucial role in the metastasis of breast cancer.
Collapse
Affiliation(s)
- Samina Malik
- Department of Physiology, University College of Medicine and Dentistry (UCMD), The University of Lahore (UOL), Lahore, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology (IMBB), UOL, Lahore, Pakistan
| | - Nimra Idrees
- Institute of Molecular Biology and Biotechnology (IMBB), UOL, Lahore, Pakistan
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), UOL, Lahore, Pakistan.
- Faculty of Health Sciences, Equator University of Science and Technology (EQUSaT), Masaka, Uganda.
| |
Collapse
|
3
|
Yang Y, Cao YL, Wang WH, Sen Shi S, Zhang YY, Lv BB, Yang WW, Li M, Wei D. Syndecan-2 modulates the YAP pathway in epithelial-to-mesenchymal transition-related migration, invasion, and drug resistance in colorectal cancer. Heliyon 2023; 9:e20183. [PMID: 37876440 PMCID: PMC10590854 DOI: 10.1016/j.heliyon.2023.e20183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/26/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is associated with an invasive phenotype in colorectal cancer (CRC). Here, we examined the roles of YES-associated protein (YAP) and syndecan-2 (SDC2) in EMT-related progression, invasion, metastasis, and drug resistance in CRC. The expression levels of YAP and SDC2 in CRC patient tumor tissue were quantified by PCR and western blotting. EMT-associated characteristics were assessed using Transwell assays and immunohistochemistry. Co-immunoprecipitation, glutathione S-transferase pull-down, and luciferase reporter assays were used to assess interactions between YAP and SDC2. YAP was found to be highly expressed in tumor tissue from 13/16 CRC patients, while SDC2 was highly expressed in the tumor tissue of 12/16 CRC patients. Overexpression of YAP in colon cancer cells led to increased cell viability, invasion, migration, and oxaliplatin resistance demonstrating that YAP plays a role in EMT. In addition, overexpression of YAP led to decreased expression of the large tumor suppressor kinase 1 (LATS1) and mammalian sterile 20-like kinases (MST1/2). Decreased LATS1 expression was associated with increased levels of cell proliferation. Knockdown of YAP by shRNA interference led to decreased cell invasion, migration, and drug resistance in colon cancer cells and reduced tumorigenesis in a mouse xenograft model. Finally, we established that YAP interacted with SDC2, and demonstrated that SDC2 mediated the YAP pathway through the EMT-related factors BMP4, CTGF and FOXM1.
Collapse
Affiliation(s)
- Yang Yang
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| | - Yong Li Cao
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| | - Wen Hang Wang
- Department of Anorectal, Zhumadian Central Hospital, Zhumadian, 463000, Henan Province, China
| | - Shou Sen Shi
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| | - Yuan Yao Zhang
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| | - Bing Bing Lv
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| | - Wei Wei Yang
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| | - Ming Li
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| | - Dong Wei
- Institute of Anal Colorectal Surgery, The 989th Hospital of the Joint Logistics Support Force of PLA, Luoyang, 471031, Henan Province, China
| |
Collapse
|
4
|
Yu D, Yang P, Lu X, Huang S, Liu L, Fan X. Single-cell RNA sequencing reveals enhanced antitumor immunity after combined application of PD-1 inhibitor and Shenmai injection in non-small cell lung cancer. Cell Commun Signal 2023; 21:169. [PMID: 37430270 DOI: 10.1186/s12964-023-01184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/04/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have altered the clinical management of non-small cell lung cancer (NSCLC). However, the low response rate, severe immune-related adverse events (irAEs), and hyperprogressive disease following ICIs monotherapy require attention. Combination therapy may overcome these limitations and traditional Chinese medicine with immunomodulatory effects provides a promising approach. Shenmai injection (SMI) is a clinically effective adjuvant treatment for cancer with chemotherapy and radiotherapy. Therefore, the combined effects and mechanisms of SMI and programmed death-1 (PD-1) inhibitor against NSCLC was focused on this study. METHODS A Lewis lung carcinoma mouse model and a lung squamous cell carcinoma humanized mouse model were used to investigate the combined efficacy and safety of SMI and PD-1 inhibitor. The synergistic mechanisms of the combination therapy against NSCLC were explored using single-cell RNA sequencing. Validation experiments were performed using immunofluorescence analysis, in vitro experiment, and bulk transcriptomic datasets. RESULTS In both models, combination therapy alleviated tumor growth and prolonged survival without increasing irAEs. The GZMAhigh and XCL1high natural killer (NK) cell subclusters with cytotoxic and chemokine signatures increased in the combination therapy, while malignant cells from combination therapy were mainly in the apoptotic state, suggesting that mediating tumor cell apoptosis through NK cells is the main synergistic mechanisms of combination therapy. In vitro experiment confirmed that combination therapy increased secretion of Granzyme A by NK cells. Moreover, we discovered that PD-1 inhibitor and SMI combination blocked inhibitory receptors on NK and T cells and restores their antitumoral activity in NSCLC better than PD-1 inhibitor monotherapy, and immune and stromal cells exhibited a decrease of angiogenic features and attenuated cancer metabolism reprogramming in microenvironment of combination therapy. CONCLUSIONS This study demonstrated that SMI reprograms tumor immune microenvironment mainly by inducing NK cells infiltration and synergizes with PD-1 inhibitor against NSCLC, suggested that targeting NK cells may be an important strategy for combining with ICIs. Video Abstract.
Collapse
Affiliation(s)
- Dingyi Yu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Penghui Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shaoze Huang
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China
| | - Li Liu
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
5
|
Puri N, Ahuja US, Gupta R, Gandhi P, Punia RS, Choudhary A. Analysis of Expression of Myofibroblasts in Oral Submucous Fibrosis: An Immunohistochemistry Study. Open Dent J 2022. [DOI: 10.2174/18742106-v16-e2201312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background:
Oral submucous fibrosis (OSMF) is a chronic disease that produces tissue fibrosis and is considered to be a potentially malignant disorder. The exact pathogenesis and malignant conversion mechanism of this disorder are still unknown. Myofibroblasts have been implicated as one of the possible pathological mechanisms responsible for the pathophysiology of OSMF. The present study was conducted to evaluate the expression of myofibroblasts (MF) in normal mucosa and different grades of OSMF.
Materials & Methods:
The sample consisted of a total of 80 specimens. The study group included specimens from clinically and histopathologically confirmed OSMF patients. The specimens were divided into four groups. Group 1 consisted of 19 specimens of grade III OSMF. Group II had 20 specimens of grade II OSMF, Group 3 with 21 specimens of grade I OSMF, and Group 4 constituted a control group of 20 normal epithelium specimens. Two sections each from all the four groups were obtained. While one section was stained with H and E, the other section was stained immunohistochemically using α-smooth muscle antibody. For analysis, the expression of myofibroblasts was categorized as strong, moderate, weak, or absent. All the results were recorded and subjected to statistical analysis.
Results:
In OSMF patients, irrespective of the grade, the expression of myofibroblast was strong in 28.33 percent of the patients, while it was moderate and weak in 30.00 percent and 40.00 percent of the patients, respectively. Expression of myofibroblast was noted to be significantly increased in grade III OSMF patients as compared to controls as well as grade I OSMF patients (p-value <0.05).
Conclusion:
Myofibroblasts expression is significantly raised in OSMF patients. The expression can also be correlated within different grades of OSMF where advanced stages show comparatively high expression of these smooth muscles like fibroblasts. Hence, we suggest that myofibroblasts could be assessed as markers for analyzing the progression of OSMF.
Collapse
|
6
|
Peng S, Hebert LL, Eschbacher JM, Kim S. Single-Cell RNA Sequencing of a Postmenopausal Normal Breast Tissue Identifies Multiple Cell Types That Contribute to Breast Cancer. Cancers (Basel) 2020; 12:cancers12123639. [PMID: 33291647 PMCID: PMC7761899 DOI: 10.3390/cancers12123639] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The human body is composed of multiple cell types that form structures and carry out the functions of specific tissues. The human breast is mainly known for the milk ducts organized by epithelial cells, but also contains many other cell types of little-known identity. In this study, we employed the single-cell sequencing technology to ascertain the various cell types present in the normal breast. The results showed 10 distinct cell types that included three epithelial and other novel cell types. The gene signatures of five cell types (three epithelial, one fibroblast subset, and immune cells) matched to the gene expression profiles of >85% breast tumors cataloged in The Cancer Gene Atlas dataset, suggesting their significant contribution to breast cancer. These findings provide a framework for the better mapping of the cellular composition in the breast and its relationship to breast disease. Abstract The human breast is composed of diverse cell types. Studies have delineated mammary epithelial cells, but the other cell types in the breast have scarcely been characterized. In order to gain insight into the cellular composition of the tissue, we performed droplet-mediated RNA sequencing of 3193 single cells isolated from a postmenopausal breast tissue without enriching for epithelial cells. Unbiased clustering analysis identified 10 distinct cell clusters, seven of which were nonepithelial devoid of cytokeratin expression. The remaining three cell clusters expressed cytokeratins (CKs), representing breast epithelial cells; Cluster 2 and Cluster 7 cells expressed luminal and basal CKs, respectively, whereas Cluster 9 cells expressed both luminal and basal CKs, as well as other CKs of unknown specificity. To assess which cell type(s) potentially contributes to breast cancer, we used the differential gene expression signature of each cell cluster to derive gene set variation analysis (GSVA) scores and classified breast tumors in The Cancer Gene Atlas (TGGA) dataset (n = 1100) by assigning the highest GSVA scoring cell cluster number for each tumor. The results showed that five clusters (Clusters 2, 3, 7, 8, and 9) could categorize >85% of breast tumors collectively. Notably, Cluster 2 (luminal epithelial) and Cluster 3 (fibroblast) tumors were equally prevalent in the luminal breast cancer subtypes, whereas Cluster 7 (basal epithelial) and Cluster 9 (other epithelial) tumors were present primarily in the triple-negative breast cancer (TNBC) subtype. Cluster 8 (immune) tumors were present in all subtypes, indicating that immune cells may contribute to breast cancer regardless of the subtypes. Cluster 9 tumors were significantly associated with poor patient survival in TNBC, suggesting that this epithelial cell type may give rise to an aggressive TNBC subset.
Collapse
Affiliation(s)
- Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA;
| | - Lora L. Hebert
- Department of Surgery, St. Joseph’s Hospital, Dignity Health, Phoenix, AZ 85013, USA; (L.L.H.); (J.M.E.)
- Surgical Breast Oncology Division, University of Arizona Cancer Center-Phoenix, Phoenix, AZ 85004, USA
| | - Jennifer M. Eschbacher
- Department of Surgery, St. Joseph’s Hospital, Dignity Health, Phoenix, AZ 85013, USA; (L.L.H.); (J.M.E.)
- Department of Neuropathology, Barrow Neurological Institute, Dignity Health, Phoenix, AZ 85013, USA
| | - Suwon Kim
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA;
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Correspondence: ; Tel.: +1-602-343-8762
| |
Collapse
|
7
|
Bahcecioglu G, Basara G, Ellis BW, Ren X, Zorlutuna P. Breast cancer models: Engineering the tumor microenvironment. Acta Biomater 2020; 106:1-21. [PMID: 32045679 PMCID: PMC7185577 DOI: 10.1016/j.actbio.2020.02.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/14/2020] [Accepted: 02/05/2020] [Indexed: 12/24/2022]
Abstract
The mechanisms behind cancer initiation and progression are not clear. Therefore, development of clinically relevant models to study cancer biology and drug response in tumors is essential. In vivo models are very valuable tools for studying cancer biology and for testing drugs; however, they often suffer from not accurately representing the clinical scenario because they lack either human cells or a functional immune system. On the other hand, two-dimensional (2D) in vitro models lack the three-dimensional (3D) network of cells and extracellular matrix (ECM) and thus do not represent the tumor microenvironment (TME). As an alternative approach, 3D models have started to gain more attention, as such models offer a platform with the ability to study cell-cell and cell-material interactions parametrically, and possibly include all the components present in the TME. Here, we first give an overview of the breast cancer TME, and then discuss the current state of the pre-clinical breast cancer models, with a focus on the engineered 3D tissue models. We also highlight two engineering approaches that we think are promising in constructing models representative of human tumors: 3D printing and microfluidics. In addition to giving basic information about the TME in the breast tissue, this review article presents the state-of-the-art tissue engineered breast cancer models. STATEMENT OF SIGNIFICANCE: Involvement of biomaterials and tissue engineering fields in cancer research enables realistic mimicry of the cell-cell and cell-extracellular matrix (ECM) interactions in the tumor microenvironment (TME), and thus creation of better models that reflect the tumor response against drugs. Engineering the 3D in vitro models also requires a good understanding of the TME. Here, an overview of the breast cancer TME is given, and the current state of the pre-clinical breast cancer models, with a focus on the engineered 3D tissue models is discussed. This review article is useful not only for biomaterials scientists aiming to engineer 3D in vitro TME models, but also for cancer researchers willing to use these models for studying cancer biology and drug testing.
Collapse
Affiliation(s)
- Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
8
|
Kim CL, Choi SH, Mo JS. Role of the Hippo Pathway in Fibrosis and Cancer. Cells 2019; 8:cells8050468. [PMID: 31100975 PMCID: PMC6562634 DOI: 10.3390/cells8050468] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
The Hippo pathway is the key player in various signaling processes, including organ development and maintenance of tissue homeostasis. This pathway comprises a core kinases module and transcriptional activation module, representing a highly conserved mechanism from Drosophila to vertebrates. The central MST1/2-LATS1/2 kinase cascade in this pathway negatively regulates YAP/TAZ transcription co-activators in a phosphorylation-dependent manner. Nuclear YAP/TAZ bind to transcription factors to stimulate gene expression, contributing to the regenerative potential and regulation of cell growth and death. Recent studies have also highlighted the potential role of Hippo pathway dysfunctions in the pathology of several diseases. Here, we review the functional characteristics of the Hippo pathway in organ fibrosis and tumorigenesis, and discuss its potential as new therapeutic targets.
Collapse
Affiliation(s)
- Cho-Long Kim
- Department of Biomedical Sciences, Cancer Biology Graduate Program, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Sue-Hee Choi
- Department of Biomedical Sciences, Cancer Biology Graduate Program, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Jung-Soon Mo
- Genomic Instability Research Center (GIRC), Ajou University School of Medicine, Suwon 16499, Korea.
| |
Collapse
|
9
|
Wadhwan V, Venkatesh A, Reddy V, Malik S. The role of myofibroblasts in the progression of oral submucous fibrosis: A systematic review. J Oral Maxillofac Pathol 2019; 23:257-266. [PMID: 31516233 PMCID: PMC6714277 DOI: 10.4103/jomfp.jomfp_238_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Oral Submucous Fibrosis (OSMF) is a chronic progressive scarring oral disease predominantly affecting people of South Asian origin. It is characterized by juxtaepithelial inflammatory cell infiltration followed by fibrosis in the lamina propria and submucosa of the oral mucosa. The pathogenesis of the disease is not well established and a number of mechanisms have been proposed regarding the pathogenesis. A renewed interest has been shown in myofibrobasts which have been implicated to play a significant role in the pathogenesis of OSMF. The myofibroblast were initially identified by means of electron microscopy in granulation tissue of healing wounds as a modulated fibroblast exhibiting features of smooth muscle cells, with prominent bundles of microfilaments, dense bodies scattered in between, and gap junctions. The presence of myofibroblasts has successively been described in practically all fibrotic situations characterized by tissue retraction and remodeling. This review paper is an attempt to identify all the studies involving myofibroblasts and explaining the pathogenesis in a simplified manner.
Collapse
Affiliation(s)
- Vijay Wadhwan
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, Subharti Dental College, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Arvind Venkatesh
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, Smile Square Multispecialty Dental Centre, Karur, Tamil Nadu, India
| | - Vandana Reddy
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, Subharti Dental College, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Sangeeta Malik
- Department of Oral Medicine and Radiology, Subharti Dental College, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| |
Collapse
|
10
|
Huliák I, Bodai L, Czepán M, Kovács D, Szabó A, Tiszlavicz L, Lázár G, Rakonczay Z, Hegyi P, Boros IM, Kiricsi M. Genetic, epigenetic and transcriptional comparison of esophagus tumor‑associated and adjacent normal myofibroblasts. Oncol Rep 2018; 41:839-852. [PMID: 30535493 PMCID: PMC6313073 DOI: 10.3892/or.2018.6909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022] Open
Abstract
Myofibroblasts (MFs) are present in healthy tissues and are also key components of the tumor microenvironment. In the present study a comparative analysis of MFs obtained from various gastrointestinal tumor tissues and from tumor-adjacent normal tissues of cancer patients was performed, with the aim to evaluate differences in MF morphology, gene expression profile and function. The goal was to correlate the observed morphological and functional variations with the underlying genetic and epigenetic backgrounds. The mutation frequency of MFs was assessed by next generation sequencing. The transcript levels of cancer-specific genes were determined by TaqMan array and quantitative polymerase chain reaction. Epigenetic modifications were analyzed by immunocytochemistry and western blotting. The migratory capacity of MFs was assessed by scratch assay, whereas matrix metalloproteinase expression and activity were obtained by quantitative polymerase chain reaction and zymography. The results of the present study demonstrate that MFs were present in an increased number and with altered morphology in tumor samples compared with the healthy tissue. Although the detected number of mutations in tumor-associated and normal tissue-derived MFs did not differ markedly, shifts in the level of specific acetylated and methylated histone proteins, namely decreased levels of trimethylated H3K9 and acetylated H4K16 were demonstrated in tumor-associated MFs. Transcript levels of several tumor-specific genes involved in metastasis, regulation of cellular growth, apoptosis, as well as in hypoxia-angiogenesis were altered in tumor-derived MF cultures. Increased mRNA levels were obtained and activity of matrix metalloproteases in tumor-derived MFs and these cells also exhibited a higher migratory capacity compared with the normal MFs. In summary, the results of the present study indicate that tumor-associated MFs display an altered phenotype compared with healthy tissue derived counterparts. The results imply that epigenetic rather than genetic alterations are associated with the development of the distinct expressional and functional features, which define this MF phenotype in the tumor microenvironment.
Collapse
Affiliation(s)
- Ildikó Huliák
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged H‑6726, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged H‑6726, Hungary
| | - Mátyás Czepán
- First Department of Medicine, University of Szeged, Szeged H‑6720, Hungary
| | - Dávid Kovács
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged H‑6726, Hungary
| | - Anikó Szabó
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged H‑6726, Hungary
| | - László Tiszlavicz
- Department of Pathology, University of Szeged, Szeged H‑6720, Hungary
| | - György Lázár
- Department of Surgery, University of Szeged, Szeged H‑6720, Hungary
| | - Zoltán Rakonczay
- First Department of Medicine, University of Szeged, Szeged H‑6720, Hungary
| | - Péter Hegyi
- First Department of Medicine, University of Szeged, Szeged H‑6720, Hungary
| | - Imre Miklós Boros
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged H‑6726, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged H‑6726, Hungary
| |
Collapse
|
11
|
Garden OA, Volk SW, Mason NJ, Perry JA. Companion animals in comparative oncology: One Medicine in action. Vet J 2018; 240:6-13. [PMID: 30268334 DOI: 10.1016/j.tvjl.2018.08.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/19/2018] [Accepted: 08/22/2018] [Indexed: 12/19/2022]
Abstract
Comparative oncology is poised to have a far-reaching impact on both animals and human beings with cancer. The field is gaining momentum and has repeatedly proven its utility in various aspects of oncology, including study of the genetics, development, progression, immunology and therapy of cancer. Companion animals provide many advantages over both traditional rodent models and human beings for studying cancer biology and accelerating the development of novel anti-cancer therapies. In this review, several examples of the ability of companion animals with spontaneous cancers to fill a unique niche in the field of oncology are discussed. In addition, potential caveats of the use of companion animals in research are reviewed, as well as ethical considerations and efforts to standardize veterinary clinical trials.
Collapse
Affiliation(s)
- O A Garden
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - S W Volk
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - N J Mason
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J A Perry
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Yazdani S, Bansal R, Prakash J. Drug targeting to myofibroblasts: Implications for fibrosis and cancer. Adv Drug Deliv Rev 2017; 121:101-116. [PMID: 28720422 DOI: 10.1016/j.addr.2017.07.010] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/20/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
Myofibroblasts are the key players in extracellular matrix remodeling, a core phenomenon in numerous devastating fibrotic diseases. Not only in organ fibrosis, but also the pivotal role of myofibroblasts in tumor progression, invasion and metastasis has recently been highlighted. Myofibroblast targeting has gained tremendous attention in order to inhibit the progression of incurable fibrotic diseases, or to limit the myofibroblast-induced tumor progression and metastasis. In this review, we outline the origin of myofibroblasts, their general characteristics and functions during fibrosis progression in three major organs: liver, kidneys and lungs as well as in cancer. We will then discuss the state-of-the art drug targeting technologies to myofibroblasts in context of the above-mentioned organs and tumor microenvironment. The overall objective of this review is therefore to advance our understanding in drug targeting to myofibroblasts, and concurrently identify opportunities and challenges for designing new strategies to develop novel diagnostics and therapeutics against fibrosis and cancer.
Collapse
Affiliation(s)
- Saleh Yazdani
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; ScarTec Therapeutics BV, Enschede, The Netherlands.
| |
Collapse
|
13
|
Comşa Ş, Popescu R, Avram Ş, Ceaușu RA, Cîmpean AM, Raica M. Bevacizumab Modulation of the Interaction Between the MCF-7 Cell Line and the Chick Embryo Chorioallantoic Membrane. In Vivo 2017; 31:199-203. [PMID: 28358700 PMCID: PMC5411745 DOI: 10.21873/invivo.11045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/17/2023]
Abstract
AIM To evaluate the interaction between MCF-7 breast cancer cells and the chick embryo chorioallantoic membrane (CAM) and the ability of bevacizumab to modulate this process. MATERIALS AND METHODS We implanted MCF-7 cells onto CAM and repeatedly added bevacizumab to a subset of eggs. We then evaluated the morphological and immunohistochemical profiles of CAM and MCF-7. RESULTS MCF-7 cells entered the mesoderm and stimulated the mesenchymal cells to acquire vasculogenic and myofibroblastoid features. MCF-7 cells developed an estrogen receptor-, progesterone receptor-, p53- and Ki67-negative status and entered the epithelial-mesenchymal transition. Bevacizumab down-regulated the expression of B-cell lymphoma 2 protein (BCL-2), vascular endothelial growth factor (VEGF) and E-cadherin in MCF-7 and inhibited vasculogenesis. CONCLUSION MCF-7 cells turn the mesoderm of CAM into a surrogate tumor stroma. CAM induces a triple-negative, non-proliferative but still anti-apoptotic status in MCF-7 cells. Although antivasculogenic, bevacizumab stimulates MCF-7 cells to acquire a more aggressive status.
Collapse
Affiliation(s)
- Şerban Comşa
- Department of Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Roxana Popescu
- Department of Cell and Molecular Biology, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Ştefana Avram
- Department of Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Raluca Amalia Ceaușu
- Department of Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Anca Maria Cîmpean
- Department of Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Marius Raica
- Department of Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| |
Collapse
|
14
|
Activation of PI3K/Akt/mTOR signaling in the tumor stroma drives endocrine therapy-dependent breast tumor regression. Oncotarget 2016; 6:22081-97. [PMID: 26098779 PMCID: PMC4673148 DOI: 10.18632/oncotarget.4203] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022] Open
Abstract
Improved efficacy of neoadjuvant endocrine-targeting therapies in luminal breast carcinomas could be achieved with optimal use of pathway targeting agents. In a mouse model of ductal breast carcinoma we identify a tumor regressive stromal reaction that is induced by neoadjuvant endocrine therapy. This reparative reaction is characterized by tumor neovascularization accompanied by infiltration of immune cells and carcinoma-associated fibroblasts that stain for phosphorylated ribosomal protein S6 (pS6), downstream the PI3K/Akt/mTOR pathway. While tumor variants with higher PI3K/Akt/mTOR activity respond well to a combination of endocrine and PI3K/Akt/mTOR inhibitors, tumor variants with lower PI3K/Akt/mTOR activity respond more poorly to the combination therapy than to the endocrine therapy alone, associated with inhibition of stromal pS6 and the reparative reaction. In human breast cancer xenografts we confirm that such differential sensitivity to therapy is primarily determined by the level of PI3K/Akt/mTOR in tumor cells. We further show that the clinical response of breast cancer patients undergoing neoadjuvant endocrine therapy is associated with the reparative stromal reaction. We conclude that tumor level and localization of pS6 are associated with therapeutic response in breast cancer and represent biomarkers to distinguish which tumors will benefit from the incorporation of PI3K/Akt/mTOR inhibitors with neoadjuvant endocrine therapy.
Collapse
|
15
|
Fong ELS, Harrington DA, Farach-Carson MC, Yu H. Heralding a new paradigm in 3D tumor modeling. Biomaterials 2016; 108:197-213. [PMID: 27639438 DOI: 10.1016/j.biomaterials.2016.08.052] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 12/14/2022]
Abstract
Numerous studies to date have contributed to a paradigm shift in modeling cancer, moving from the traditional two-dimensional culture system to three-dimensional (3D) culture systems for cancer cell culture. This led to the inception of tumor engineering, which has undergone rapid advances over the years. In line with the recognition that tumors are not merely masses of proliferating cancer cells but rather, highly complex tissues consisting of a dynamic extracellular matrix together with stromal, immune and endothelial cells, significant efforts have been made to better recapitulate the tumor microenvironment in 3D. These approaches include the development of engineered matrices and co-cultures to replicate the complexity of tumor-stroma interactions in vitro. However, the tumor engineering and cancer biology fields have traditionally relied heavily on the use of cancer cell lines as a cell source in tumor modeling. While cancer cell lines have contributed to a wealth of knowledge in cancer biology, the use of this cell source is increasingly perceived as a major contributing factor to the dismal failure rate of oncology drugs in drug development. Backing this notion is the increasing evidence that tumors possess intrinsic heterogeneity, which predominantly homogeneous cancer cell lines poorly reflect. Tumor heterogeneity contributes to therapeutic resistance in patients. To overcome this limitation, cancer cell lines are beginning to be replaced by primary tumor cell sources, in the form of patient-derived xenografts and organoids cultures. Moving forward, we propose that further advances in tumor engineering would require that tumor heterogeneity (tumor variants) be taken into consideration together with tumor complexity (tumor-stroma interactions). In this review, we provide a comprehensive overview of what has been achieved in recapitulating tumor complexity, and discuss the importance of incorporating tumor heterogeneity into 3D in vitro tumor models. This work carves out the roadmap for 3D tumor engineering and highlights some of the challenges that need to be addressed as we move forward into the next chapter.
Collapse
Affiliation(s)
- Eliza L S Fong
- Department of Physiology, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore.
| | | | | | - Hanry Yu
- Department of Physiology, National University of Singapore, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research, Singapore; Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Guo Z, Zhang T, Fang K, Liu P, Li M, Gu N. The effect of porosity and stiffness of glutaraldehyde cross-linked egg white scaffold simulating aged extracellular matrix on distribution and aggregation of ovarian cancer cells. Colloids Surf A Physicochem Eng Asp 2016. [DOI: 10.1016/j.colsurfa.2016.05.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
17
|
Marsh T, Wong I, Sceneay J, Barakat A, Qin Y, Sjödin A, Alspach E, Nilsson B, Stewart SA, McAllister SS. Hematopoietic Age at Onset of Triple-Negative Breast Cancer Dictates Disease Aggressiveness and Progression. Cancer Res 2016; 76:2932-43. [PMID: 27197230 DOI: 10.1158/0008-5472.can-15-3332] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/18/2016] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is considered an early onset subtype of breast cancer that carries with it a poorer prognosis in young rather than older women for reasons that remain poorly understood. Hematopoiesis in the bone marrow becomes altered with age and may therefore affect the composition of tumor-infiltrating hematopoietic cells and subsequent tumor progression. In this study, we investigated how age- and tumor-dependent changes to bone marrow-derived hematopoietic cells impact TNBC progression. Using multiple mouse models of TNBC tumorigenesis and metastasis, we found that a specific population of bone marrow cells (BMC) upregulated CSF-1R and secreted the growth factor granulin to support stromal activation and robust tumor growth in young mice. However, the same cell population in old mice expressed low levels of CSF1R and granulin and failed to promote tumor outgrowth, suggesting that age influences the tumorigenic capacity of BMCs in response to tumor-associated signals. Importantly, BMCs from young mice were sufficient to activate a tumor-supportive microenvironment and induce tumor progression in old mice. These results indicate that hematopoietic age is an important determinant of TNBC aggressiveness and provide rationale for investigating age-stratified therapies designed to prevent the protumorigenic effects of activated BMCs. Cancer Res; 76(10); 2932-43. ©2016 AACR.
Collapse
Affiliation(s)
- Timothy Marsh
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Irene Wong
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jaclyn Sceneay
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Amey Barakat
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yuanbo Qin
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Andreas Sjödin
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
| | - Elise Alspach
- Department of Cell Biology and Physiology; Department of Medicine; and ICCE Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Björn Nilsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Sheila A Stewart
- Department of Cell Biology and Physiology; Department of Medicine; and ICCE Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Sandra S McAllister
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts. Harvard Stem Cell Institute, Cambridge, Massachusetts.
| |
Collapse
|
18
|
Relationship between ultrasound elastography and myofibroblast distribution in breast cancer and its clinical significance. Sci Rep 2016; 6:19584. [PMID: 26846996 PMCID: PMC4742804 DOI: 10.1038/srep19584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 07/10/2015] [Indexed: 12/16/2022] Open
Abstract
The study investigated the relationship between ultrasound elastography (USE) scoring and myofibroblast distribution with expression features of α-SMA + /CD34− in patients of Uyghur and Han ethnicities with breast masses in Xinjiang, China. The data was used to evaluate its clinical significance in the early diagnosis of breast cancer. A total of 300 patients with breast masses were included in the study, which involved conventional sonography and USE, with histopathologic diagnosis as the reference standard. Myofibroblast distribution was investigated by detecting the expression levels of α-SMA and CD34 in lesions using immunohistochemistry and real-time PCR. Out of 300 lesions, 185 were histologically malignant and 115 benign. The mean elasticity score for malignant lesions was significantly higher than for benign lesions. The expression level of α-SMA was elevated while the expression level of CD34 was lower in malignancies, compared with benign lesions. The expression of α-SMA was positively associated with the USE scores, while a negative relationship was observed between CD34 expression and USE scoring. The combination of USE and molecular diagnosis provides a promising modality for the early diagnosis and evaluation of the risks in particular types of breast cancer.
Collapse
|
19
|
Kolahian S, Öz HH, Zhou B, Griessinger CM, Rieber N, Hartl D. The emerging role of myeloid-derived suppressor cells in lung diseases. Eur Respir J 2016; 47:967-77. [PMID: 26846830 DOI: 10.1183/13993003.01572-2015] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/15/2015] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are innate immune cells characterised by their potential to control T-cell responses and to dampen inflammation. While the role of MDSCs in cancer has been studied in depth, our understanding of their relevance for infectious and inflammatory disease conditions has just begun to evolve. Recent studies highlight an emerging and complex role for MDSCs in pulmonary diseases. In this review, we discuss the potential contribution of MDSCs as biomarkers and therapeutic targets in lung diseases, particularly lung cancer, tuberculosis, chronic obstructive pulmonary disease, asthma and cystic fibrosis.
Collapse
Affiliation(s)
- Saeed Kolahian
- Children's Hospital of the University of Tübingen, Pediatric Infectiology, Immunology & Cystic Fibrosis, Tübingen, Germany Dept of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Hasan Halit Öz
- Children's Hospital of the University of Tübingen, Pediatric Infectiology, Immunology & Cystic Fibrosis, Tübingen, Germany
| | - Benyuan Zhou
- Children's Hospital of the University of Tübingen, Pediatric Infectiology, Immunology & Cystic Fibrosis, Tübingen, Germany
| | - Christoph M Griessinger
- Werner Siemens Imaging Center, Dept of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nikolaus Rieber
- Children's Hospital of the University of Tübingen, Pediatric Infectiology, Immunology & Cystic Fibrosis, Tübingen, Germany Dept of Pediatrics, Kinderklinik München Schwabing, Klinikum rechts der Isar, Technische Universität München, Munich Germany
| | - Dominik Hartl
- Children's Hospital of the University of Tübingen, Pediatric Infectiology, Immunology & Cystic Fibrosis, Tübingen, Germany
| |
Collapse
|
20
|
Veiseh M, Leith SJ, Tolg C, Elhayek SS, Bahrami SB, Collis L, Hamilton S, McCarthy JB, Bissell MJ, Turley E. Uncovering the dual role of RHAMM as an HA receptor and a regulator of CD44 expression in RHAMM-expressing mesenchymal progenitor cells. Front Cell Dev Biol 2015; 3:63. [PMID: 26528478 PMCID: PMC4606125 DOI: 10.3389/fcell.2015.00063] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/17/2015] [Indexed: 12/31/2022] Open
Abstract
The interaction of hyaluronan (HA) with mesenchymal progenitor cells impacts trafficking and fate after tissue colonization during wound repair and these events contribute to diseases such as cancer. How this interaction occurs is poorly understood. Using 10T½ cells as a mesenchymal progenitor model and fluorescent (F-HA) or gold-labeled HA (G-HA) polymers, we studied the role of two HA receptors, RHAMM and CD44, in HA binding and uptake in non-adherent and adherent mesenchymal progenitor (10T½) cells to mimic aspects of cell trafficking and tissue colonization. We show that fluorescent labeled HA (F-HA) binding/uptake was high in non-adherent cells but dropped over time as cells became increasingly adherent. Non-adherent cells displayed both CD44 and RHAMM but only function-blocking anti-RHAMM and not anti-CD44 antibodies significantly reduced F-HA binding/uptake. Adherent cells, which also expressed CD44 and RHAMM, primarily utilized CD44 to bind to F-HA since anti-CD44 but not anti-RHAMM antibodies blocked F-HA uptake. RHAMM overexpression in adherent 10T½ cells led to increased F-HA uptake but this increased binding remained CD44 dependent. Further studies showed that RHAMM-transfection increased CD44 mRNA and protein expression while blocking RHAMM function reduced expression. Collectively, these results suggest that cellular microenvironments in which these receptors function as HA binding proteins differ significantly, and that RHAMM plays at least two roles in F-HA binding by acting as an HA receptor in non-attached cells and by regulating CD44 expression and display in attached cells. Our findings demonstrate adhesion-dependent mechanisms governing HA binding/ uptake that may impact development of new mesenchymal cell-based therapies.
Collapse
Affiliation(s)
- Mandana Veiseh
- Life Sciences Division, Lawrence Berkeley National Laboratories Berkeley, CA, USA ; Palo Alto Research Center (a Xerox Company) Palo Alto, CA, USA
| | - Sean J Leith
- Departments of Oncology/Biochemistry/Surgery, Western Schulich School of Medicine, London Regional Cancer Program, Western University London, ON, Canada
| | - Cornelia Tolg
- Departments of Oncology/Biochemistry/Surgery, Western Schulich School of Medicine, London Regional Cancer Program, Western University London, ON, Canada
| | - Sallie S Elhayek
- Departments of Oncology/Biochemistry/Surgery, Western Schulich School of Medicine, London Regional Cancer Program, Western University London, ON, Canada
| | - S Bahram Bahrami
- Life Sciences Division, Lawrence Berkeley National Laboratories Berkeley, CA, USA
| | - Lisa Collis
- Departments of Oncology/Biochemistry/Surgery, Western Schulich School of Medicine, London Regional Cancer Program, Western University London, ON, Canada
| | - Sara Hamilton
- Departments of Oncology/Biochemistry/Surgery, Western Schulich School of Medicine, London Regional Cancer Program, Western University London, ON, Canada
| | - James B McCarthy
- Department of Laboratory Medicine and Pathology, Masonic Comprehensive Cancer Center, University of Minnesota Minneapolis, MN, USA
| | - Mina J Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratories Berkeley, CA, USA
| | - Eva Turley
- Departments of Oncology/Biochemistry/Surgery, Western Schulich School of Medicine, London Regional Cancer Program, Western University London, ON, Canada
| |
Collapse
|
21
|
Chen X, Lu P, Chen L, Yang SJ, Shen HY, Yu DD, Zhang XH, Zhong SL, Zhao JH, Tang JH. Perioperative propofol-paravertebral anesthesia decreases the metastasis and progression of breast cancer. Tumour Biol 2015; 36:8259-66. [PMID: 26383520 DOI: 10.1007/s13277-015-4027-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 08/31/2015] [Indexed: 01/29/2023] Open
Abstract
Propofol-paravertebral anesthesia (PPA) is a unique combination of paravertebral nerve blocks (PVBs) and propofol that regulates the cellular microenvironment during surgical period. Growing evidence points to its ability to attenuate perioperative immunosuppression of cancers. Abundant studies show that cancer patients who undergo perioperative PPA exhibit less recurrence as well as metastasis. Breast cancer remains a leading cause of cancer-induced death in women. Over the last decades, increasing concerns have been put on the promotional role of PPA in the prognosis of breast cancer patients. Among them, PPA participates in several bioprocesses in the development of breast cancer, including inhibiting hypoxia-inducible factor (HIF) activity, elevating serum concentration of nitric oxide index (NOx), depression of the neuroepithelial cell transforming gene 1 (NET1) signal pathway, blocking the nuclear factor kappa B (NF-κB) pathway following an decreased expression of matrix metalloproteinase (MMP), increasing NK cytotoxicity, and affecting transforming growth factor (TGF)-β-targeted ras and HER2/neu gene pathways. In this review, we discuss the effect of PPA on breast cancer metastasis and progression. This will provide an alteration pattern of surgical anesthesia technique in breast cancer patients with poor prognosis.
Collapse
Affiliation(s)
- Xiu Chen
- The Fourth Clinical School of Nanjing Medical University, Baiziting 42, Nanjing, 210009, China.,Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Peng Lu
- School of Public Healthy Nanjing Medical University, Jiangsulu 172, Nanjing, 210009, China
| | - Lin Chen
- Department of Oncology, Xuzhou Medical College, Xuzhou, 221004, China
| | - Su-jin Yang
- The Fourth Clinical School of Nanjing Medical University, Baiziting 42, Nanjing, 210009, China.,Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Hong-Yu Shen
- The Fourth Clinical School of Nanjing Medical University, Baiziting 42, Nanjing, 210009, China.,Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Dan-dan Yu
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Xiao-hui Zhang
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Shan-liang Zhong
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China
| | - Jian-hua Zhao
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China.
| | - Jin-hai Tang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Baiziting 42, Nanjing, 210009, China.
| |
Collapse
|
22
|
Cichon MA, Radisky DC. Extracellular matrix as a contextual determinant of transforming growth factor-β signaling in epithelial-mesenchymal transition and in cancer. Cell Adh Migr 2015; 8:588-94. [PMID: 25482625 PMCID: PMC4594483 DOI: 10.4161/19336918.2014.972788] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Extracellular matrix (ECM) provides both structural support and contextual information to cells within tissues and organs. The combination of biochemical and biomechanical signals from the ECM modulates responses to extracellular signals toward differentiation, proliferation, or apoptosis; alterations in the ECM are necessary for development and remodeling processes, but aberrations in the composition and organization of ECM are associated with disease pathology and can predispose to development of cancer. The primary cell surface sensors of the ECM are the integrins, which provide the physical connection between the ECM and the cytoskeleton and also convey biochemical information about the composition of the ECM. Transforming growth factor-β (TGF-β) is an extracellular signaling molecule that is a powerful controller of a variety of cellular functions, and that has been found to induce very different outcomes according to cell type and cellular context. It is becoming clear that ECM-mediated signaling through integrins is reciprocally influenced by TGF-β: integrin expression, activation, and responses are affected by cellular exposure to TGF-β, and TGF-β activation and cellular responses are in turn controlled by signaling from the ECM through integrins. Epithelial-mesenchymal transition (EMT), a physiological process that is activated by TGF-β in normal development and in cancer, is also affected by the composition and structure of the ECM. Here, we will outline how signaling from the ECM controls the contextual response to TGF-β, and how this response is selectively modulated during disease, with an emphasis on recent findings, current challenges, and future opportunities.
Collapse
|
23
|
Radisky ES, Radisky DC. Matrix metalloproteinases as breast cancer drivers and therapeutic targets. Front Biosci (Landmark Ed) 2015; 20:1144-63. [PMID: 25961550 DOI: 10.2741/4364] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Members of the matrix metalloproteinase (MMP) family have been identified as poor prognosis markers for breast cancer patients and as drivers of many facets of the tumor phenotype in experimental models. Early enthusiasm for MMPs as therapeutic targets was tempered following disappointing clinical trials that utilized broad spectrum, small molecule catalytic site inhibitors. However, subsequent research has continued to define key roles for MMPs as breast cancer promoters, to elucidate the complex roles that that these proteins play in breast cancer development and progression, and to identify how these roles are linked to specific and unique biochemical features of individual members of the MMP family. Here, we provide an overview of the structural features of the MMPs, then discuss clinical studies identifying which MMP family members are linked with breast cancer development and new experimental studies that reveal how these specific MMPs may play unique roles in the breast cancer microenvironment. We conclude with a discussion of the most promising avenues for development of therapeutic agents capable of targeting the tumor-promoting properties of MMPs.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224,
| | | |
Collapse
|
24
|
Primary Myofibroblasts Maintain Short-Term Viability following Submucosal Injection in Syngeneic, Immune-Competent Mice Utilizing Murine Colonoscopy. PLoS One 2015; 10:e0127258. [PMID: 26016485 PMCID: PMC4445916 DOI: 10.1371/journal.pone.0127258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 04/13/2015] [Indexed: 01/24/2023] Open
Abstract
The myofibroblast is an important stromal cell of the gastrointestinal tract. Current in vitro and in vivo models either do not accurately recreate stromal-epithelial interactions or are not specific to myofibroblasts. We sought to create an animal model that would allow the study of myofibroblast-epithelial interactions. We isolated and cultured colonic myofibroblasts from FVB mice. Cells were α-SMA and vimentin positive but desmin negative on immunoblot analysis. We injected the myofibroblasts into the colonic submucosa of syngeneic adult mice (n = 8) via a miniendoscopic system. We then isolated green fluorescent protein (GFP) positive colonic myofibroblasts from C57BL/6-Tg(CAG-EGFP)1Osb/J mice and injected them into the colonic lamina propria of C57BL/6J mice at 1x105 (n = 14), 1x106 (n = 9), or 5x106 cells/mL (n = 4). A subset of mice were injected with serum-free media and ink without cells (n = 3). Mice underwent repeat endoscopy and euthanasia one or 7 days after injection. Colons were isolated and either fixed in 10% formalin or the inked sites were individually excised and lysed for DNA. We assessed the injection sites via histology and immunohistochemical stains for α-SMA and GFP. We used qPCR to quantify GFP DNA transcripts at the lamina propria injection sites. Submucosal injection of myofibroblasts resulted in the formation of a subepithelial wheal on endoscopy, which persisted to day 7. Myofibroblasts injected either into the submucosa or lamina propria maintained viability on post-injection day 7 as evidenced by positive α-SMA staining. qPCR of lamina propria injections showed a dose-dependent increase in GFP DNA transcripts on post-injection day 1, whereas the number of transcripts on day 7 was equivalent for the concentrations injected. We demonstrate short-term survival of primary cultured colonic myofibroblasts in syngeneic mice. This may prove to be a valuable model for studying the role of myofibroblasts in states of health and disease.
Collapse
|
25
|
Brisson BK, Mauldin EA, Lei W, Vogel LK, Power AM, Lo A, Dopkin D, Khanna C, Wells RG, Puré E, Volk SW. Type III Collagen Directs Stromal Organization and Limits Metastasis in a Murine Model of Breast Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1471-86. [PMID: 25795282 DOI: 10.1016/j.ajpath.2015.01.029] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/31/2014] [Accepted: 01/22/2015] [Indexed: 01/02/2023]
Abstract
Breast cancer metastasis is the leading cause of cancer-related deaths in women worldwide. Collagen in the tumor microenvironment plays a crucial role in regulating tumor progression. We have shown that type III collagen (Col3), a component of tumor stroma, regulates myofibroblast differentiation and scar formation after cutaneous injury. During the course of these wound-healing studies, we noted that tumors developed at a higher frequency in Col3(+/-) mice compared to wild-type littermate controls. We, therefore, examined the effect of Col3 deficiency on tumor behavior, using the murine mammary carcinoma cell line 4T1. Notably, tumor volume and pulmonary metastatic burden after orthotopic injection of 4T1 cells were increased in Col3(+/-) mice compared to Col3(+/+) littermates. By using murine (4T1) and human (MDA-MB-231) breast cancer cells grown in Col3-poor and Col3-enriched microenvironments in vitro, we found that several major events of the metastatic process were suppressed by Col3, including adhesion, invasion, and migration. In addition, Col3 deficiency increased proliferation and decreased apoptosis of 4T1 cells both in vitro and in primary tumors in vivo. Mechanistically, Col3 suppresses the procarcinogenic microenvironment by regulating stromal organization, including density and alignment of fibrillar collagen and myofibroblasts. We propose that Col3 plays an important role in the tumor microenvironment by suppressing metastasis-promoting characteristics of the tumor-associated stroma.
Collapse
Affiliation(s)
- Becky K Brisson
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth A Mauldin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Weiwei Lei
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laurie K Vogel
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ashley M Power
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Albert Lo
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Derek Dopkin
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chand Khanna
- Tumor and Metastasis Biology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Rebecca G Wells
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan W Volk
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
26
|
Becker MA, Haluska P, Bale LK, Oxvig C, Conover CA. A novel neutralizing antibody targeting pregnancy-associated plasma protein-a inhibits ovarian cancer growth and ascites accumulation in patient mouse tumorgrafts. Mol Cancer Ther 2015; 14:973-81. [PMID: 25695953 DOI: 10.1158/1535-7163.mct-14-0880] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/10/2015] [Indexed: 12/16/2022]
Abstract
The majority of ovarian cancer patients acquire resistance to standard platinum chemotherapy and novel therapies to reduce tumor burden and ascites accumulation are needed. Pregnancy-associated plasma protein-A (PAPP-A) plays a key role in promoting insulin-like growth factor (IGF) pathway activity, which directly correlates to ovarian cancer cell transformation, growth, and invasiveness. Herein, we evaluate PAPP-A expression in tumors and ascites of women with ovarian cancer, and determine the antitumor efficacy of a neutralizing monoclonal PAPP-A antibody (mAb-PA) in ovarian cancer using primary patient ovarian tumorgrafts ("Ovatars"). PAPP-A mRNA expression in patient ovarian tumors correlated with poor outcome and was validated as a prognostic surrogate in Ovatar tumors. Following confirmation of mAb-PA bioavailability and target efficacy in vivo, the antitumor efficacy of mAb-PA in multiple Ovatar tumor models was examined and the response was found to depend on PAPP-A expression. Strikingly, the addition of mAb-PA to standard platinum chemotherapy effectively sensitized platinum-resistant Ovatar tumors. PAPP-A protein in ascites was also assessed in a large cohort of patients and very high levels were evident across the entire sample set. Therefore, we evaluated targeted PAPP-A inhibition as a novel approach to managing ovarian ascites, and found that mAb-PA inhibited the development, attenuated the progression, and induced the regression of Ovatar ascites. Together, these data indicate PAPP-A as a potential palliative and adjunct therapeutic target for women with ovarian cancer.
Collapse
Affiliation(s)
- Marc A Becker
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Paul Haluska
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Laurie K Bale
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
27
|
Cichon MA, Nelson CM, Radisky DC. Regulation of epithelial-mesenchymal transition in breast cancer cells by cell contact and adhesion. Cancer Inform 2015; 14:1-13. [PMID: 25698877 PMCID: PMC4325704 DOI: 10.4137/cin.s18965] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/29/2014] [Accepted: 01/04/2015] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a physiological program that is activated during cancer cell invasion and metastasis. We show here that EMT-related processes are linked to a broad and conserved program of transcriptional alterations that are influenced by cell contact and adhesion. Using cultured human breast cancer and mouse mammary epithelial cells, we find that reduced cell density, conditions under which cell contact is reduced, leads to reduced expression of genes associated with mammary epithelial cell differentiation and increased expression of genes associated with breast cancer. We further find that treatment of cells with matrix metalloproteinase-3 (MMP-3), an inducer of EMT, interrupts a defined subset of cell contact-regulated genes, including genes encoding a variety of RNA splicing proteins known to regulate the expression of Rac1b, an activated splice isoform of Rac1 known to be a key mediator of MMP-3-induced EMT in breast, lung, and pancreas. These results provide new insights into how MMPs act in cancer progression and how loss of cell-cell interactions is a key step in the earliest stages of cancer development.
Collapse
Affiliation(s)
- Magdalena A Cichon
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL USA
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL USA
| |
Collapse
|
28
|
Reyes-Uribe E, Serna-Marquez N, Perez Salazar E. DDRs: receptors that mediate adhesion, migration and invasion in breast cancer cells. AIMS BIOPHYSICS 2015. [DOI: 10.3934/biophy.2015.3.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
29
|
Deficiency of Kruppel-like factor KLF4 in myeloid-derived suppressor cells inhibits tumor pulmonary metastasis in mice accompanied by decreased fibrocytes. Oncogenesis 2014; 3:e129. [PMID: 25417726 PMCID: PMC4259966 DOI: 10.1038/oncsis.2014.44] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/15/2014] [Indexed: 01/20/2023] Open
Abstract
The importance of immunosuppressive myeloid-derived suppressor cells (MDSCs) bearing monocyte markers in tumor metastasis has been well established. Recently, it was reported that these cells possess phenotypic plasticity and differentiate into fibrocytes, very distinct cells that are precursors of tumorigenic myofibroblasts. However, the importance of this transdifferentiation in tumor metastasis has not been explored. Here, we describe the role of MDSC-derived fibrocytes in tumor metastasis that is regulated by Kruppel-like factor 4 (KLF4), a transcription factor that is critical to monocyte differentiation and to promotion of cancer development. Using mouse metastasis models of melanoma and breast cancer, we found that KLF4 knockout was associated with significantly reduced pulmonary metastasis, which was accompanied by decreased populations of MDSCs, fibrocytes and myofibroblasts in the lung. Cause-effect studies by adoptive transfer revealed that KLF4 deficiency in MDSCs led to significantly reduced lung metastasis that was associated with fewer MDSC-derived fibrocytes and myofibroblasts. Mechanistically, KLF4 deficiency significantly compromised the generation of fibrocytes from MDSCs in vitro. During this process, KLF4 expression levels were tightly linked with those of fibroblast-specific protein-1 (FSP-1), deficiency of which resulted in no metastasis in mice as has been previously reported. In addition, KLF4 bound directly to the FSP-1 promoter as determined by chromatin immunoprecipitation and overexpression of KLF4 increased the FSP-1 promoter activities. Taken together, our results suggest that MDSCs not only execute their immunosuppressive function to promote metastatic seeding as reported before, but also boost metastatic tumor growth after they adopt a fibrocyte fate. Therefore, KLF4-mediated fibrocyte generation from MDSCs may represent a novel mechanism of MDSCs contributing to tumor metastasis and supports the feasibility of inhibiting KLF4 or FSP-1 to prevent tumor metastasis.
Collapse
|
30
|
Mehner C, Miller E, Khauv D, Nassar A, Oberg AL, Bamlet WR, Zhang L, Waldmann J, Radisky ES, Crawford HC, Radisky DC. Tumor cell-derived MMP3 orchestrates Rac1b and tissue alterations that promote pancreatic adenocarcinoma. Mol Cancer Res 2014; 12:1430-9. [PMID: 24850902 PMCID: PMC4201965 DOI: 10.1158/1541-7786.mcr-13-0557-t] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
UNLABELLED Pancreatic ductal adenocarcinoma (PDA) arises at the convergence of genetic alterations in KRAS with a fostering microenvironment shaped by immune cell influx and fibrotic changes; identification of the earliest tumorigenic molecular mediators evokes the proverbial chicken and egg problem. Matrix metalloproteinases (MMP) are key drivers of tumor progression that originate primarily from stromal cells activated by the developing tumor. Here, MMP3, known to be expressed in PDA, was found to be associated with expression of Rac1b, a tumorigenic splice isoform of Rac1, in all stages of pancreatic cancer. Using a large cohort of human PDA tissue biopsies specimens, both MMP3 and Rac1b are expressed in PDA cells, that the expression levels of the two markers are highly correlated, and that the subcellular distribution of Rac1b in PDA is significantly associated with patient outcome. Using transgenic mouse models, coexpression of MMP3 with activated KRAS in pancreatic acinar cells stimulates metaplasia and immune cell infiltration, priming the stromal microenvironment for early tumor development. Finally, exposure of cultured pancreatic cancer cells to recombinant MMP3 stimulates expression of Rac1b, increases cellular invasiveness, and activation of tumorigenic transcriptional profiles. IMPLICATIONS MMP3 acts as a coconspirator of oncogenic KRAS in pancreatic cancer tumorigenesis and progression, both through Rac1b-mediated phenotypic control of pancreatic cancer cells themselves, and by giving rise to the tumorigenic microenvironment; these findings also point to inhibition of this pathway as a potential therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Christine Mehner
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 U S A
| | - Erin Miller
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 U S A
| | - Davitte Khauv
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 U S A
| | - Aziza Nassar
- Department of Pathology, Mayo Clinic, Jacksonville, Florida
| | - Ann L Oberg
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - William R Bamlet
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research
| | - Lizhi Zhang
- Department of Pathology, Mayo Clinic, Rochester, Minnesota; and
| | - Jens Waldmann
- Department of Visceral-, Thoracic- and Vascular Surgery, Unikliniken Marburg Und Giessen, Marburg, Germany
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 U S A
| | - Howard C Crawford
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 U S A
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 U S A;
| |
Collapse
|
31
|
Translation in solid cancer: are size-based response criteria an anachronism? Clin Transl Oncol 2014; 17:1-10. [PMID: 25073600 DOI: 10.1007/s12094-014-1207-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 07/09/2014] [Indexed: 12/19/2022]
Abstract
The purpose of translation is the development of effective medicinal products based on validated science. A parallel objective is to obtain marketing authorization for the translated product. Unfortunately, in solid cancer, these two objectives are not mutually consistent as evidenced by the contrast between major advances in science and the continuing dismal record of pharmaceutical productivity. If the problem is unrelated to science, then the process of translation may require a closer examination, namely, the criteria for regulatory approval. This realization is important because, in this context, the objective of translation is regulatory approval, and science does not passively translate into useful medicinal products. Today, in solid cancer, response criteria related to tumor size are less useful than during the earlier cytotoxic drugs era; advanced imaging and biomarkers now allow for tracking of the natural history of the disease in the laboratory and the clinic. Also, it is difficult to infer clinical benefit from tumor shrinkage since it is rarely sustained. Accordingly, size-based response criteria may represent an anachronism relative to translation in solid cancer and it may be appropriate to align preclinical and clinical effort and shift the focus to local invasion and metastasis. The shift from a cancer cell-centric model to a stroma centric model offers novel opportunities not only to interupt the natural history of the disease, but also to rethink the relevance of outdated criteria of clinical response. Current evidence favors the opinion that, in solid cancer, a different, broader, and contextual approach may lead to interventions that could delay local invasion and metastasis. All elements supporting this shift, especially advanced imaging, are in place.
Collapse
|
32
|
Håkanson M, Cukierman E, Charnley M. Miniaturized pre-clinical cancer models as research and diagnostic tools. Adv Drug Deliv Rev 2014; 69-70:52-66. [PMID: 24295904 PMCID: PMC4019677 DOI: 10.1016/j.addr.2013.11.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/09/2013] [Accepted: 11/24/2013] [Indexed: 12/14/2022]
Abstract
Cancer is one of the most common causes of death worldwide. Consequently, important resources are directed towards bettering treatments and outcomes. Cancer is difficult to treat due to its heterogeneity, plasticity and frequent drug resistance. New treatment strategies should strive for personalized approaches. These should target neoplastic and/or activated microenvironmental heterogeneity and plasticity without triggering resistance and spare host cells. In this review, the putative use of increasingly physiologically relevant microfabricated cell-culturing systems intended for drug development is discussed. There are two main reasons for the use of miniaturized systems. First, scaling down model size allows for high control of microenvironmental cues enabling more predictive outcomes. Second, miniaturization reduces reagent consumption, thus facilitating combinatorial approaches with little effort and enables the application of scarce materials, such as patient-derived samples. This review aims to give an overview of the state-of-the-art of such systems while predicting their application in cancer drug development.
Collapse
Affiliation(s)
- Maria Håkanson
- CSEM SA, Section for Micro-Diagnostics, 7302 Landquart, Switzerland
| | - Edna Cukierman
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | - Mirren Charnley
- Centre for Micro-Photonics and Industrial Research Institute Swinburne, Swinburne University of Technology, Victoria 3122, Australia.
| |
Collapse
|