1
|
Mundhara N, Sadhukhan P. Cracking the Codes behind Cancer Cells' Immune Evasion. Int J Mol Sci 2024; 25:8899. [PMID: 39201585 PMCID: PMC11354234 DOI: 10.3390/ijms25168899] [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: 07/12/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Immune evasion is a key phenomenon in understanding tumor recurrence, metastasis, and other critical steps in tumor progression. The tumor microenvironment (TME) is in constant flux due to the tumor's ability to release signals that affect it, while immune cells within it can impact cancer cell behavior. Cancer cells undergo several changes, which can change the enrichment of different immune cells and modulate the activity of existing immune cells in the tumor microenvironment. Cancer cells can evade immune surveillance by downregulating antigen presentation or expressing immune checkpoint molecules. High levels of tumor-infiltrating lymphocytes (TILs) correlate with better outcomes, and robust immune responses can control tumor growth. On the contrary, increased enrichment of Tregs, myeloid-derived suppressor cells, and M2-like anti-inflammatory macrophages can hinder effective immune surveillance and predict poor prognosis. Overall, understanding these immune evasion mechanisms guides therapeutic strategies. Researchers aim to modulate the TME to enhance immune surveillance and improve patient outcomes. In this review article, we strive to summarize the composition of the tumor immune microenvironment, factors affecting the tumor immune microenvironment (TIME), and different therapeutic modalities targeting the immune cells. This review is a first-hand reference to understand the basics of immune surveillance and immune evasion.
Collapse
Affiliation(s)
| | - Pritam Sadhukhan
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
2
|
Ocadiz-Ruiz R, Decker JT, Griffin K, Tan ZM, Domala NK, Jeruss JS, Shea LD. Human Breast Cancer Cell Lines Differentially Modulate Signaling from Distant Microenvironments, Which Reflects Their Metastatic Potential. Cancers (Basel) 2024; 16:796. [PMID: 38398186 PMCID: PMC10887178 DOI: 10.3390/cancers16040796] [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: 11/21/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Metastasis is the stage at which the prognosis substantially decreases for many types of cancer. The ability of tumor cells to metastasize is dependent upon the characteristics of the tumor cells, and the conditioning of distant tissues that support colonization by metastatic cells. In this report, we investigated the systemic alterations in distant tissues caused by multiple human breast cancer cell lines and the impact of these alterations on the tumor cell phenotype. We observed that the niche within the lung, a common metastatic site, was significantly altered by MDA-MB-231, MCF7, and T47 tumors, and that the lung microenvironment stimulated, to differing extents, an epithelial-to-mesenchymal transition (EMT), reducing proliferation, increasing transendothelial migration and senescence, with no significant impact on cell death. We also investigated the ability of an implantable scaffold, which supports the formation of a distant tissue, to serve as a surrogate for the lung to identify systemic alterations. The scaffolds are conditioned by the primary tumor similarly to the lung for each tumor type, evidenced by promoting a pro-EMT profile. Collectively, we demonstrate that metastatic and non-metastatic breast cancers condition distant tissues, with distinct effects on tumor cell responses, and that a surrogate tissue can distinguish the metastatic potential of human breast cancer cell lines in an accessible site that avoids biopsy of a vital organ.
Collapse
Affiliation(s)
- Ramon Ocadiz-Ruiz
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.O.-R.)
| | - Joseph T. Decker
- Department of Cariology, Restorative Sciences, and Endodontics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kate Griffin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.O.-R.)
| | - Zoey M. Tan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.O.-R.)
| | - Nishant K. Domala
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.O.-R.)
| | - Jacqueline S. Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.O.-R.)
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.O.-R.)
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Zivari-Ghader T, Dolati S, Mehdizadeh A, Davaran S, Rashidi MR, Yousefi M. Recent scaffold-based tissue engineering approaches in premature ovarian failure treatment. J Tissue Eng Regen Med 2022; 16:605-620. [PMID: 35511799 DOI: 10.1002/term.3306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022]
Abstract
Recently, tissue engineering and regenerative medicine have received significant attention with outstanding advances. The main scope of this technology is to recover the damaged tissues and organs or to maintain and improve their function. One of the essential fields in tissue engineering is scaffold designing and construction, playing an integral role in damaged tissues reconstruction and repair. However, premature ovarian failure (POF) is a disorder causing many medical and psychological problems in women. POF treatment using tissue engineering and various scaffold has recently made tremendous and promising progress. Due to the importance of the subject, we have summarized the recently examined scaffolds in the treatment of POF in this review.
Collapse
Affiliation(s)
- Tayyebeh Zivari-Ghader
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Guo L, Liu M, Dou Y, Duan R, Shen L, Jia L, Wang J, Li C, Li X, Liang T. Screening and identification of haptoglobin showing its important role in pathophysiological process of gallbladder carcinoma. Gene 2021; 776:145429. [PMID: 33444685 DOI: 10.1016/j.gene.2021.145429] [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: 09/21/2020] [Revised: 12/17/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
Gallbladder cancer (GBC) with poor prognosis has been a major cause of cancer-related deaths worldwide. In this study, we aimed to screen and identify crucial genes in GBC through integrative analysis of multiple datasets and further experimental validation. A candidate crucial gene, up-regulated haptoglobin (HP), was firstly screened, and then further analysis and validation mainly focused on whether higher enrichment level of HP was responsible for pathophysiological process of GBC. HP was found with diverse expression patterns in various cancer types, and the dynamic expression patterns indicated its spatiotemporal characteristics in different tissues and disease stages, implicating its role in multiple biological processes. Further experimental validation showed that HP could promote the GBC-SD cell proliferation, migration and invasion, implying its role in pathophysiological process of GBC. HP may have a crucial role in occurrence and development of GBC, and it provides possibility as a potential biomarker or target in cancer prognosis and treatment.
Collapse
Affiliation(s)
- Li Guo
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Mengting Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Yuyang Dou
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Rui Duan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Lulu Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Lin Jia
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China
| | - Jun Wang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Changxian Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiangcheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, and Changzhou Institute of Innovation and Development, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
5
|
Wang WY, Jarman EH, Lin D, Baker BM. Dynamic Endothelial Stalk Cell-Matrix Interactions Regulate Angiogenic Sprout Diameter. Front Bioeng Biotechnol 2021; 9:620128. [PMID: 33869150 PMCID: PMC8044977 DOI: 10.3389/fbioe.2021.620128] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is a complex, multicellular process that involves bidirectional interactions between extracellular matrix (ECM) and collectively invading endothelial cell (EC) sprouts that extend the microvasculature during development, wound healing, and disease processes. While many aspects of angiogenesis have been well studied, the relationship between endothelial sprout morphology and subsequent neovessel function remains relatively unknown. Here, we investigated how various soluble and physical matrix cues that regulate endothelial sprouting speed and proliferation correspond to changes in sprout morphology, namely, sprout stalk diameter. We found that sprout stalk cells utilize a combination of cytoskeletal forces and proteolysis to physically compact and degrade the surrounding matrix, thus creating sufficient space in three-dimensional (3D) ECM for lateral expansion. As increasing sprout diameter precedes lumenization to generate perfusable neovessels, this work highlights how dynamic endothelial stalk cell-ECM interactions promote the generation of functional neovessels during sprouting angiogenesis to provide insight into the design of vascularized, implantable biomaterials.
Collapse
Affiliation(s)
| | | | | | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
6
|
Garg SK, Welsh EA, Fang B, Hernandez YI, Rose T, Gray J, Koomen JM, Berglund A, Mulé JJ, Markowitz J. Multi-Omics and Informatics Analysis of FFPE Tissues Derived from Melanoma Patients with Long/Short Responses to Anti-PD1 Therapy Reveals Pathways of Response. Cancers (Basel) 2020; 12:cancers12123515. [PMID: 33255891 PMCID: PMC7768436 DOI: 10.3390/cancers12123515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/21/2020] [Indexed: 01/14/2023] Open
Abstract
Simple Summary Immune based therapies have benefited many melanoma patients, but many patients still do not respond. This study analyzes biospecimens obtained from patients undergoing a type of immune based therapy called anti-PD-1 to understand mechanisms of response and resistance to this treatment. The operational definition of good response utilized in this investigation permitted us to examine the biochemical pathways that are facilitating anti-PD-1 responses independent of prior therapies received by patients. Currently, there are no clinically available tests to reliably test for the outcome of patients treated with anti-PD-1 therapy. The purpose of this study was to facilitate the development of prospective biomarker-directed trials to guide therapy, as even though the side effect profile is favorable for anti-PD-1 therapy, some patients do not respond to therapy with significant toxicity. Each patient may require testing for the pathways upregulated in the tumor to predict optimal benefit to anti-PD-1 treatment. Abstract Anti-PD-1 based immune therapies are thought to be dependent on antigen processing and presentation mechanisms. To characterize the immune-dependent mechanisms that predispose stage III/IV melanoma patients to respond to anti-PD-1 therapies, we performed a multi-omics study consisting of expression proteomics and targeted immune-oncology-based mRNA sequencing. Formalin-fixed paraffin-embedded tissue samples were obtained from stage III/IV patients with melanoma prior to anti-PD-1 therapy. The patients were first stratified into poor and good responders based on whether their tumors had or had not progressed while on anti-PD-1 therapy for 1 year. We identified 263 protein/gene candidates that displayed differential expression, of which 223 were identified via proteomics and 40 via targeted-mRNA analyses. The downstream analyses of expression profiles using MetaCore software demonstrated an enrichment of immune system pathways involved in antigen processing/presentation and cytokine production/signaling. Pathway analyses showed interferon (IFN)-γ-mediated signaling via NF-κB and JAK/STAT pathways to affect immune processes in a cell-specific manner and to interact with the inducible nitric oxide synthase. We review these findings within the context of available literature on the efficacy of anti-PD-1 therapy. The comparison of good and poor responders, using efficacy of PD-1-based therapy at 1 year, elucidated the role of antigen presentation in mediating response or resistance to anti-PD-1 blockade.
Collapse
Affiliation(s)
- Saurabh K. Garg
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (S.K.G.); (Y.I.H.)
| | - Eric A. Welsh
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Bin Fang
- Proteomics & Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (B.F.); (J.M.K.)
| | - Yuliana I. Hernandez
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (S.K.G.); (Y.I.H.)
| | - Trevor Rose
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
- Department of Oncologic Sciences, University of South Florida Health Morsani College of Medicine, Tampa, FL 33620, USA; (J.G.); (A.B.); (J.J.M.)
| | - Jhanelle Gray
- Department of Oncologic Sciences, University of South Florida Health Morsani College of Medicine, Tampa, FL 33620, USA; (J.G.); (A.B.); (J.J.M.)
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M. Koomen
- Proteomics & Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (B.F.); (J.M.K.)
- Department of Oncologic Sciences, University of South Florida Health Morsani College of Medicine, Tampa, FL 33620, USA; (J.G.); (A.B.); (J.J.M.)
| | - Anders Berglund
- Department of Oncologic Sciences, University of South Florida Health Morsani College of Medicine, Tampa, FL 33620, USA; (J.G.); (A.B.); (J.J.M.)
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - James J. Mulé
- Department of Oncologic Sciences, University of South Florida Health Morsani College of Medicine, Tampa, FL 33620, USA; (J.G.); (A.B.); (J.J.M.)
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Joseph Markowitz
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (S.K.G.); (Y.I.H.)
- Department of Oncologic Sciences, University of South Florida Health Morsani College of Medicine, Tampa, FL 33620, USA; (J.G.); (A.B.); (J.J.M.)
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-745-8581
| |
Collapse
|
7
|
Morris AH, Orbach SM, Bushnell GG, Oakes RS, Jeruss JS, Shea LD. Engineered Niches to Analyze Mechanisms of Metastasis and Guide Precision Medicine. Cancer Res 2020; 80:3786-3794. [PMID: 32409307 PMCID: PMC7501202 DOI: 10.1158/0008-5472.can-20-0079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/04/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
Cancer metastasis poses a challenging problem both clinically and scientifically, as the stochastic nature of metastatic lesion formation introduces complexity for both early detection and the study of metastasis in preclinical models. Engineered metastatic niches represent an emerging approach to address this stochasticity by creating bioengineered sites where cancer can preferentially metastasize. As the engineered niche captures the earliest metastatic cells at a nonvital location, both noninvasive and biopsy-based monitoring of these sites can be performed routinely to detect metastasis early and monitor alterations in the forming metastatic niche. The engineered metastatic niche also provides a new platform technology that serves as a tunable site to molecularly dissect metastatic disease mechanisms. Ultimately, linking the engineered niches with advances in sensor development and synthetic biology can provide enabling tools for preclinical cancer models and fosters the potential to impact the future of clinical cancer care.
Collapse
Affiliation(s)
- Aaron H Morris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Sophia M Orbach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Robert S Oakes
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Jacqueline S Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
8
|
Aguado BA, Schuetze KB, Grim JC, Walker CJ, Cox AC, Ceccato TL, Tan AC, Sucharov CC, Leinwand LA, Taylor MRG, McKinsey TA, Anseth KS. Transcatheter aortic valve replacements alter circulating serum factors to mediate myofibroblast deactivation. Sci Transl Med 2020; 11:11/509/eaav3233. [PMID: 31511425 PMCID: PMC6754739 DOI: 10.1126/scitranslmed.aav3233] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/25/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022]
Abstract
The transcatheter aortic valve replacement (TAVR) procedure has emerged as a minimally invasive treatment for patients with aortic valve stenosis (AVS). However, alterations in serum factor composition and biological activity after TAVR remain unknown. Here, we quantified the systemic inflammatory effects of the TAVR procedure and hypothesized that alterations in serum factor composition would modulate valve and cardiac fibrosis. Serum samples were obtained from patients with AVS immediately before their TAVR procedure (pre-TAVR) and about 1 month afterward (post-TAVR). Aptamer-based proteomic profiling revealed alterations in post-TAVR serum composition, and ontological analysis identified inflammatory macrophage factors implicated in myofibroblast activation and deactivation. Hydrogel biomaterials used as valve matrix mimics demonstrated that post-TAVR serum reduced myofibroblast activation of valvular interstitial cells relative to pre-TAVR serum from the same patient. Transcriptomics and curated network analysis revealed a shift in myofibroblast phenotype from pre-TAVR to post-TAVR and identified p38 MAPK signaling as one pathway involved in pre-TAVR–mediated myofibroblast activation. Post-TAVR serum deactivated valve and cardiac myofibroblasts initially exposed to pre-TAVR serum to a quiescent fibroblast phenotype. Our in vitro deactivation data correlated with patient disease severity measured via echocardiography and multimorbidity scores, and correlations were dependent on hydrogel stiffness. Sex differences in cellular responses to male and female sera were also observed and may corroborate clinical observations regarding sex-specific TAVR outcomes. Together, alterations in serum composition after TAVR may lead to an antifibrotic fibroblast phenotype, which suggests earlier interventions may be beneficial for patients with advanced AVS to prevent further disease progression. Transcatheter aortic valve replacement alters a patient’s serum proteome, reversing valvular interstitial cell and cardiac myofibroblast activation. Aortic valve stenosis (narrowing of the aortic valve) contributes to inadequate blood flow, fibrosis, hypertrophy, and, ultimately, heart failure. Transcatheter aortic valve replacement (TAVR) improves blood flow, but little is known about cardiac remodeling after the procedure. Aguado and colleagues performed proteomics on serum samples collected from patients before and after TAVR and studied the effects of serum on valve and cardiac cells using hydrogel culture platforms. A role for p38 MAPK signaling in activating cells was identified using pre-TAVR serum, whereas post-TAVR serum returned cells to a quiescent state. Along with preliminary insights into sex-specific differences, the authors’ research supports a role for TAVR-induced alteration of circulating inflammatory cytokines in regulating valve cell phenotype.
Collapse
Affiliation(s)
- Brian A Aguado
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Katherine B Schuetze
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joseph C Grim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Cierra J Walker
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA.,Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Anne C Cox
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Tova L Ceccato
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA.,Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Aik-Choon Tan
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Leslie A Leinwand
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA.,Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Matthew R G Taylor
- Department of Medicine, Adult Clinical Genetics, University of Colorado Health Science Center, Aurora, CO 80045, USA
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA. .,Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA. .,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
9
|
Bushnell GG, Hong X, Hartfield RM, Zhang Y, Oakes RS, Rao SS, Jeruss JS, Stegemann JP, Deng CX, Shea LD. High Frequency Spectral Ultrasound Imaging to Detect Metastasis in Implanted Biomaterial Scaffolds. Ann Biomed Eng 2020; 48:477-489. [PMID: 31549327 PMCID: PMC6930322 DOI: 10.1007/s10439-019-02366-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022]
Abstract
For most cancers, metastasis is the point at which disease is no longer curable. Earlier detection of metastasis, when it is undetectable by current clinical methods, may enable better outcomes. We have developed a biomaterial implant that recruits metastatic cancer cells in mouse models of breast cancer. Here, we investigate spectral ultrasound imaging (SUSI) as a non-invasive strategy for detecting metastasis to the implanted biomaterial scaffolds. Our results show that SUSI, which detects parameters related to tissue composition and structure, identified changes at an early time point when tumor cells were recruited to scaffolds in orthotopic breast cancer mouse models. These changes were not associated with acellular components in the scaffolds but were reflected in the cellular composition in the scaffold microenvironment, including an increase in CD31 + CD45-endothelial cell number in tumor bearing mice. In addition, we built a classification model based on changes in SUSI parameters from scaffold measurements to stratify tumor free and tumor bearing status. Combination of a linear discriminant analysis and bagged decision trees model resulted in an area under the curve of 0.92 for receiver operating characteristics analysis. With the potential for early non-invasive detection, SUSI could facilitate clinical translation of the scaffolds for monitoring metastatic disease.
Collapse
Affiliation(s)
- Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiaowei Hong
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rachel M Hartfield
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yining Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert S Oakes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Jacqueline S Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Cheri X Deng
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI, 48109-2099, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, 2111 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI, 48109-2099, USA.
| |
Collapse
|
10
|
Bushnell GG, Rao SS, Hartfield RM, Zhang Y, Oakes RS, Jeruss JS, Shea LD. Microporous scaffolds loaded with immunomodulatory lentivirus to study the contribution of immune cell populations to tumor cell recruitment in vivo. Biotechnol Bioeng 2020; 117:210-222. [PMID: 31544959 PMCID: PMC6991704 DOI: 10.1002/bit.27179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/23/2019] [Accepted: 09/13/2019] [Indexed: 01/13/2023]
Abstract
Metastases are preceded by stochastic formation of a hospitable microenvironment known as the premetastatic niche, which has been difficult to study. Herein, we employ implantable polycaprolactone scaffolds as an engineered premetastatic niche to independently investigate the role of interleukin-10 (IL10), CXCL12, and CCL2 in recruiting immune and tumor cells and impacting breast cancer cell phenotype via lentiviral overexpression. Lentivirus delivered from scaffolds in vivo achieved sustained transgene expression for 56 days. IL10 lentiviral expression, but not CXCL12 or CCL2, significantly decreased tumor cell recruitment to scaffolds in vivo. Delivery of CXCL12 enhanced CD45+ immune cell recruitment to scaffolds while delivery of IL10 reduced immune cell recruitment. CCL2 did not alter immune cell recruitment. Tumor cell phenotype was investigated using conditioned media from immunomodulated scaffolds, with CXCL12 microenvironments reducing proliferation, and IL10 microenvironments enhancing proliferation. Migration was enhanced with CCL2 and reduced with IL10-driven microenvironments. Multiple linear regression identified populations of immune cells associated with tumor cell abundance. CD45+ immune and CD8+ T cells were associated with reduced tumor cell abundance, while CD11b+Gr1+ neutrophils and CD4+ T cells were associated with enhanced tumor cell abundance. Collectively, biomaterial scaffolds provide a tool to probe the formation and function of the premetastatic niche.
Collapse
Affiliation(s)
- Grace G. Bushnell
- Department of Biomedical Engineering, University of
Michigan, Ann Arbor, Michigan
| | - Shreyas S. Rao
- Department of Chemical and Biological Engineering,
University of Alabama, Tuscaloosa, Alabama
| | - Rachel M. Hartfield
- Department of Biomedical Engineering, University of
Michigan, Ann Arbor, Michigan
| | - Yining Zhang
- Department of Chemical Engineering, University of Michigan,
Ann Arbor, Michigan
| | - Robert S. Oakes
- Department of Biomedical Engineering, University of
Michigan, Ann Arbor, Michigan
| | - Jacqueline S. Jeruss
- Department of Biomedical Engineering, University of
Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor,
Michigan
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of
Michigan, Ann Arbor, Michigan
- Department of Chemical Engineering, University of Michigan,
Ann Arbor, Michigan
| |
Collapse
|
11
|
Lee HR, Pelaez F, Silbaugh AM, Leslie F, Racila E, Azarin SM. Biomaterial Platform To Establish a Hypoxic Metastatic Niche in Vivo. ACS APPLIED BIO MATERIALS 2019; 2:1549-1560. [PMID: 35026889 DOI: 10.1021/acsabm.8b00837] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxia is a hallmark of tumor microenvironments, exerting wide-ranging impacts on key processes of tumor progression and metastasis. However, our understanding of how hypoxia regulates these processes has been based primarily on studying the effects of hypoxia within the primary tumor. Recently, an increasing number of studies have suggested the importance of hypoxic regulation within metastatic target organs, but hypoxic metastatic niches in the body are difficult to access with current imaging techniques, hampering detailed in vivo investigation of hypoxia at metastatic sites. Here, we report an engineered biomaterial scaffold that is able to establish an in vivo hypoxic metastatic niche in a readily accessible area, enabling the investigation of hypoxic regulation at a metastatic site. We engineered hypoxic environments within microporous poly(lactide-co-glycolide) (PLG) scaffolds, which have previously been shown to act as premetastatic niche mimics, via the addition of CoCl2, a hypoxia-mimetic agent. When implanted into the subcutaneous region of mice, CoCl2-containing PLG (Co-PLG) scaffolds established hypoxic microenvironments, as evidenced by the stabilization of hypoxia-inducible factor 1α (HIF1α) and increased blood vessel formation in vitro and in vivo. Furthermore, implanted Co-PLG scaffolds were able to recruit 4T1 metastatic breast cancer cells. These results demonstrate that Co-PLG scaffolds can establish an in vivo hypoxic metastatic niche, providing a novel platform to investigate hypoxic regulation of disseminated tumor cells (DTCs) at target organs.
Collapse
|
12
|
Bushnell GG, Hardas TP, Hartfield RM, Zhang Y, Oakes RS, Ronquist S, Chen H, Rajapakse I, Wicha MS, Jeruss JS, Shea LD. Biomaterial Scaffolds Recruit an Aggressive Population of Metastatic Tumor Cells In Vivo. Cancer Res 2019; 79:2042-2053. [PMID: 30808673 PMCID: PMC6467791 DOI: 10.1158/0008-5472.can-18-2502] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/21/2018] [Accepted: 02/18/2019] [Indexed: 12/28/2022]
Abstract
For most cancers, metastasis is the point at which clinical treatment shifts from curative intent to extending survival. Biomaterial implants acting as a synthetic premetastatic niche recruit metastatic cancer cells and provide a survival advantage, and their use as a diagnostic platform requires assessing their relevance to disease progression. Here, we showed that scaffold-captured tumor cells (SCAF) were 30 times more metastatic to the lung than primary tumor (PT) cells, similar to cells derived from lung micrometastases (LUNG). SCAF cells were more aggressive in vitro, demonstrated higher levels of migration, invasion, and mammosphere formation, and had a greater proportion of cancer stem cells than PT. SCAF cells were highly enriched for gene expression signatures associated with metastasis and had associated genomic structural changes, including globally enhanced entropy. Collectively, our findings demonstrate that SCAF cells are distinct from PT and more closely resemble LUNG, indicating that tumor cells retrieved from scaffolds are reflective of cells at metastatic sites. SIGNIFICANCE: These findings suggest that metastatic tumor cells captured by a biomaterial scaffold may serve as a diagnostic for molecular staging of metastasis.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/8/2042/F1.large.jpg.
Collapse
Affiliation(s)
- Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Tejaswini P Hardas
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Rachel M Hartfield
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Yining Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Robert S Oakes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Scott Ronquist
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Haiming Chen
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Indika Rajapakse
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jacqueline S Jeruss
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
13
|
Carpenter RA, Kwak JG, Peyton SR, Lee J. Implantable pre-metastatic niches for the study of the microenvironmental regulation of disseminated human tumour cells. Nat Biomed Eng 2018; 2:915-929. [PMID: 30906645 PMCID: PMC6424369 DOI: 10.1038/s41551-018-0307-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 09/06/2018] [Indexed: 02/07/2023]
Abstract
Cancer survivors often carry disseminated tumour cells (DTCs), yet owing to DTC dormancy they do not relapse from treatment. Understanding how the local microenvironment regulates the transition of DTCs from a quiescent state to active proliferation could suggest new therapeutic strategies to prevent or delay the formation of metastases. Here, we show that implantable biomaterial microenvironments incorporating human stromal cells, immune cells and cancer cells can be used to examine the post-dissemination phase of the evolution of the tumour microenvironment. After subdermal implantation in mice, porous hydrogel scaffolds seeded with human bone marrow stromal cells form a vascularized niche and recruit human circulating tumour cells released from an orthotopic prostate tumour xenograft. Systemic injection of human peripheral blood mononuclear cells slowed the evolution of the active metastatic niches but did not change the rate of overt metastases, as the ensuing inflammation promoted the formation of DTC colonies. Implantable pre-metastatic niches might enable the study of DTC colonization and proliferation, and facilitate the development of effective anti-metastatic therapies.
Collapse
Affiliation(s)
- Ryan A Carpenter
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Jun-Goo Kwak
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA.
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
14
|
Beri P, Matte BF, Fattet L, Kim D, Yang J, Engler AJ. Biomaterials to model and measure epithelial cancers. NATURE REVIEWS. MATERIALS 2018; 3:418-430. [PMID: 30416759 PMCID: PMC6224155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The use of biomaterials has substantially contributed to both our understanding of tumorigenesis and our ability to identify and capture tumour cells in vitro and in vivo. Natural and synthetic biomaterials can be applied as models to recapitulate key features of the tumour microenvironment in vitro, including architectural, mechanical and biological functions. Engineered biomaterials can further mimic the spatial and temporal properties of the surrounding tumour niche to investigate the specific effects of the environment on disease progression, offering an alternative to animal models for the testing of cancer cell behaviour. Biomaterials can also be used to capture and detect cancer cells in vitro and in vivo to monitor tumour progression. In this Review, we discuss the natural and synthetic biomaterials that can be used to recreate specific features of tumour microenvironments. We examine how biomaterials can be applied to capture circulating tumour cells in blood samples for the early detection of metastasis. We highlight biomaterial-based strategies to investigate local regions adjacent to the tumour and survey potential applications of biomaterial-based devices for diagnosis and prognosis, such as the detection of cellular deformability and the non-invasive surveillance of tumour-adjacent stroma.
Collapse
Affiliation(s)
- Pranjali Beri
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Bibiana F. Matte
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Department of Oral Pathology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Laurent Fattet
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Daehwan Kim
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jing Yang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Adam J. Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| |
Collapse
|
15
|
Synergy of Paracrine Signaling During Early-Stage Mouse Ovarian Follicle Development In Vitro. Cell Mol Bioeng 2018; 11:435-450. [PMID: 31719893 DOI: 10.1007/s12195-018-0545-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/20/2018] [Indexed: 10/28/2022] Open
Abstract
Introduction Paracrine signals, such as soluble cytokines and extracellular matrix cues, are essential for the survival and development of multicellular ovarian follicles. While it is well established that hydrogel-based culture systems successfully support the growth of late-stage follicles for fertility preservation, growing small, early-stage ovarian follicles still proves to be challenging. We hypothesized that paracrine factors secreted from neighboring follicles may be crucial for improving the survival of early-stage follicles in vitro. Methods To test our hypothesis, we investigated the bi-directional crosstalk of the paracrine signals, such as cell-secreted cytokines, sex hormones and transcription factors (TFs), in follicles encapsulated and cultured for 12 days in alginate in groups of five (5×) and ten (10×). Results The differential profiles of TF activity and secretome during folliculogenesis were analyzed using TRanscriptional Activity CEllular aRray (TRACER) and data-driven multivariate modeling approach. The mechano- and oxygen-responsive TFs, NF-κB and HIF1, exhibited a unique upregulation signature in 10× follicles. Consistently, levels of proangiogenic factors, such as VEGF-A and angiopoietin-2, were significantly higher in 10× follicles than those in 5× follicles, reaching 269.77 and 242.82 pg/mL on the last day of culture. The analysis of TRACER and secreted cytokines also revealed critical early interactions between cytokines and TFs, correlating with the observed phenotypical and functional differences between conditions. Conclusions We identified unique signatures of synergism during successful early-stage ovarian follicle development. These findings bring us closer to understanding of mechanisms underlying the downstream effects of interactions between the extracellular microenvironment and early-stage folliculogenesis in vitro.
Collapse
|
16
|
Aguado BA, Hartfield RM, Bushnell GG, Decker JT, Azarin SM, Nanavati D, Schipma MJ, Rao SS, Oakes RS, Zhang Y, Jeruss JS, Shea LD. Biomaterial Scaffolds as Pre-metastatic Niche Mimics Systemically Alter the Primary Tumor and Tumor Microenvironment. Adv Healthc Mater 2018; 7:e1700903. [PMID: 29521008 PMCID: PMC6014830 DOI: 10.1002/adhm.201700903] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 01/28/2018] [Indexed: 12/11/2022]
Abstract
Primary tumor (PT) immune cells and pre-metastatic niche (PMN) sites are critical to metastasis. Recently, synthetic biomaterial scaffolds used as PMN mimics are shown to capture both immune and metastatic tumor cells. Herein, studies are performed to investigate whether the scaffold-mediated redirection of immune and tumor cells would alter the primary tumor microenvironment (TME). Transcriptomic analysis of PT cells from scaffold-implanted and mock-surgery mice identifies differentially regulated pathways relevant to invasion and metastasis progression. Transcriptomic differences are hypothesized to result from scaffold-mediated modulations of immune cell trafficking and phenotype in the TME. Culturing tumor cells with conditioned media generated from PT immune cells of scaffold-implanted mice decrease invasion in vitro more than two-fold relative to mock surgery controls and reduce activity of invasion-promoting transcription factors. Secretomic characterization of the conditioned media delineates interactions between immune cells in the TME and tumor cells, showing an increase in the pan-metastasis inhibitor decorin and a concomitant decrease in invasion-promoting chemokine (C-C motif) ligand 2 (CCL2) in scaffold-implanted mice. Flow cytometric and transcriptomic profiling of PT immune cells identify phenotypically distinct tumor-associated macrophages (TAMs) in scaffold-implanted mice, which may contribute to an invasion-suppressive TME. Taken together, this study demonstrates biomaterial scaffolds systemically influence metastatic progression through manipulation of the TME.
Collapse
Affiliation(s)
- Brian A Aguado
- Department of Biomedical Engineering, Simpson Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
| | - Rachel M Hartfield
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Dhaval Nanavati
- Proteomics Core Facility, Northwestern University, Chicago, IL, 60611, USA
| | - Matthew J Schipma
- NUSeq Core Facility, Northwestern University, Chicago, IL, 60611, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Robert S Oakes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Yining Zhang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| | | | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| |
Collapse
|
17
|
Abstract
The pre-metastatic niche — the accumulation of aberrant immune cells and extracellular matrix proteins in target organs — primes the initially healthy organ microenvironment and renders it amenable for subsequent metastatic cell colonization. By attracting metastatic cancer cells, mimics of the pre-metastatic niche offer both diagnostic and therapeutic potential. However, deconstructing the complexity of the niche by identifying the interactions between cell populations and the mediatory roles of the immune system, soluble factors, extracellular matrix proteins, and stromal cells has proved challenging. Experimental models need to recapitulate niche-population biology in situ and mediate in vivo tumour-cell homing, colonization and proliferation. In this Review, we outline the biology of the pre-metastatic niche and discuss advances in engineered niche-mimicking biomaterials that regulate the behaviour of tumour cells at an implant site. Such oncomaterials offer strategies for early detection of metastatic events, inhibiting the formation of the pre-metastatic niche, and attenuating metastatic progression.
Collapse
|
18
|
Bowden M. The Cancer Secretome. CANCER DRUG DISCOVERY AND DEVELOPMENT 2017:95-120. [DOI: 10.1007/978-3-319-45397-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Schulten HJ, Hussein D, Al-Adwani F, Karim S, Al-Maghrabi J, Al-Sharif M, Jamal A, Bakhashab S, Weaver J, Al-Ghamdi F, Baeesa SS, Bangash M, Chaudhary A, Al-Qahtani M. Microarray expression profiling identifies genes, including cytokines, and biofunctions, as diapedesis, associated with a brain metastasis from a papillary thyroid carcinoma. Am J Cancer Res 2016; 6:2140-2161. [PMID: 27822408 PMCID: PMC5088282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 06/06/2023] Open
Abstract
Brain metastatic papillary thyroid carcinomas (PTCs) are afflicted with unfavorable prognosis; however, the underlying molecular genetics of these rare metastases are virtually unknown. In this study, we compared whole transcript microarray expression profiles of a BRAF mutant, brain metastasis from a PTC, including its technical replicate (TR), with eight non-brain metastatic PTCs and eight primary brain tumors. The top 95 probe sets (false discovery rate (FDR) p-value < 0.05 and fold change (FC) > 2) that were differentially expressed between the brain metastatic PTC, including the TR, and both, non-brain metastatic PTCs and primary brain tumors were in the vast majority upregulated and comprise, e.g. ROS1, MYBPH, SLC18A3, HP, SAA2-SAA4, CP, CCL20, GFAP, RNU1-120P, DMBT1, XDH, CXCL1, PI3, and NAPSA. Cytokines were represented by 10 members in the top 95 probe sets. Pathway and network analysis (p-value < 0.05 and FC > 2) identified granulocytes adhesion and diapedesis as top canonical pathway. Most significant upstream regulators were lipopolysaccharide, TNF, NKkB (complex), IL1A, and CSF2. Top networks categorized under diseases & functions were entitled migration of cells, cell movement, cell survival, apoptosis, and proliferation of cells. Probe sets that were significantly shared between the brain metastatic PTC, the TR, and primary brain tumors include CASP1, CASP4, C1R, CC2D2B, RNY1P16, WDR72, LRRC2, ZHX2, CITED1, and the noncoding transcript AK128523. Taken together, this study identified a set of candidate genes and biofunctions implicated in, so far nearly uncharacterized, molecular processes of a brain metastasis from a PTC.
Collapse
Affiliation(s)
- Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, King Abdulaziz UniversityJeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Deema Hussein
- King Fahad Medical Research Center, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Fatima Al-Adwani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz UniversityJeddah, Saudi Arabia
- Department of Biology, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research, King Abdulaziz UniversityJeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Jaudah Al-Maghrabi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University HospitalJeddah, Saudi Arabia
- Department of Pathology, King Faisal Specialist Hospital and Research CenterJeddah, Saudi Arabia
| | - Mona Al-Sharif
- Department of Biology, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Awatif Jamal
- Department of Pathology, Faculty of Medicine, King Abdulaziz University HospitalJeddah, Saudi Arabia
| | - Sherin Bakhashab
- Center of Excellence in Genomic Medicine Research, King Abdulaziz UniversityJeddah, Saudi Arabia
- Institute of Cellular Medicine, Newcastle UniversityNewcastle NE2 4HH, United Kingdom
| | - Jolanta Weaver
- Institute of Cellular Medicine, Newcastle UniversityNewcastle NE2 4HH, United Kingdom
| | - Fahad Al-Ghamdi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University HospitalJeddah, Saudi Arabia
| | - Saleh S Baeesa
- Division of Neurosurgery, Department of Surgery, King Abdulaziz University HospitalJeddah, Saudi Arabia
| | - Mohammed Bangash
- Division of Neurosurgery, Department of Surgery, King Abdulaziz University HospitalJeddah, Saudi Arabia
| | - Adeel Chaudhary
- Center of Excellence in Genomic Medicine Research, King Abdulaziz UniversityJeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Mohammed Al-Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz UniversityJeddah, Saudi Arabia
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz UniversityJeddah, Saudi Arabia
| |
Collapse
|
20
|
Weng TY, Wang CY, Hung YH, Chen WC, Chen YL, Lai MD. Differential Expression Pattern of THBS1 and THBS2 in Lung Cancer: Clinical Outcome and a Systematic-Analysis of Microarray Databases. PLoS One 2016; 11:e0161007. [PMID: 27513329 PMCID: PMC4981437 DOI: 10.1371/journal.pone.0161007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022] Open
Abstract
Thrombospondin 1 and thrombospondin 2 (THBS1 and THBS2) share similar multifunctional domains, and are known to be antiangiogenic. However, the expression pattern of THBS1 and THBS2 is different, and the specific role of THBS2 in different subtypes of lung cancer remains largely unclear. To evaluate the significance of THBS1 and THBS2 in the development of lung cancer, the present study performed a microarray-based systematic-analysis to determine the transcript levels of thrombospondins and their relation to the prognosis in lung cancer. THBS1 was in general underexpressed in lung cancer; in contrast, mRNA levels of THBS2 were markedly overexpressed in a number of datasets of non-small cell lung carcinoma (NSCLC), including lung adenocarcinoma (AC) and squamous cell carcinoma. Similar expression pattern of THBS1 and THBS2 was verified in pulmonary AC cell lines with real-time PCR analysis. The survival of lung AC patients with high THBS2 mRNA expression levels was poorer than patients with low levels of expression of THBS2. In a microarray-based analysis, genes coexpressed with THBS1 or THBS2 were determined. Pulmonary AC patients with a high expression level of sevenTSHB1-coexpressed genes (CCL5, CDH11, FYB, GZMK, LA-DQA1, PDE4DIP, and SELL) had better survival rates than those with a low expression level. Patients with a high expression of seven TSHB2-coexpressed genes (CHI3L1, COL5A2, COL11A1, FAP, MXRA5, THY1, and VCAN) had poor survival rates. Downregulation of VCAN and THBS2 with shRNA inhibited the cell proliferation in the A549 cell line. In summary, THBS1 functions as a tumor suppressor in lung adenocarcinoma. However, THBS2 may play a double-edged role in the progression of lung AC, i.e. anti-angiogenic and oncogenic function. Further study on the mechanism underlying the activity of THBS2 is warranted to have further implications for cancer diagnosis and treatment of pulmonary AC.
Collapse
Affiliation(s)
- Tzu-Yang Weng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Yang Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hsuan Hung
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Ching Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ling Chen
- Department of Senior Citizen Services Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
21
|
Aguado BA, Caffe JR, Nanavati D, Rao SS, Bushnell GG, Azarin SM, Shea LD. Extracellular matrix mediators of metastatic cell colonization characterized using scaffold mimics of the pre-metastatic niche. Acta Biomater 2016; 33:13-24. [PMID: 26844426 PMCID: PMC4777643 DOI: 10.1016/j.actbio.2016.01.043] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/22/2015] [Accepted: 01/30/2016] [Indexed: 12/12/2022]
Abstract
Metastatic tumor cells colonize the pre-metastatic niche, which is a complex microenvironment consisting partially of extracellular matrix (ECM) proteins. We sought to identify and validate novel contributors to tumor cell colonization using ECM-coated poly(ε-caprolactone) (PCL) scaffolds as mimics of the pre-metastatic niche. Utilizing orthotopic breast cancer mouse models, fibronectin and collagen IV-coated scaffolds implanted in the subcutaneous space captured colonizing tumor cells, showing a greater than 2-fold increase in tumor cell accumulation at the implant site compared to uncoated scaffolds. As a strategy to identify additional ECM colonization contributors, decellularized matrix (DCM) from lungs and livers containing metastatic tumors were characterized. In vitro, metastatic cell adhesion was increased on DCM coatings from diseased organs relative to healthy DCM. Furthermore, in vivo implantations of diseased DCM-coated scaffolds had increased tumor cell colonization relative to healthy DCM coatings. Mass-spectrometry proteomics was performed on healthy and diseased DCM to identify candidates associated with colonization. Myeloperoxidase was identified as abundantly present in diseased organs and validated as a contributor to colonization using myeloperoxidase-coated scaffold implants. This work identified novel ECM proteins associated with colonization using decellularization and proteomics techniques and validated candidates using a scaffold to mimic the pre-metastatic niche. STATEMENT OF SIGNIFICANCE The pre-metastatic niche consists partially of ECM proteins that promote metastatic cell colonization to a target organ. We present a biomaterials-based approach to mimic this niche and identify ECM mediators of colonization. Using murine breast cancer models, we implanted microporous PCL scaffolds to recruit colonizing tumor cells in vivo. As a strategy to modulate colonization, we coated scaffolds with various ECM proteins, including decellularized lung and liver matrix from tumor-bearing mice. After characterizing the organ matrices using proteomics, myeloperoxidase was identified as an ECM protein contributing to colonization and validated using our scaffold. Our scaffold provides a platform to identify novel contributors to colonization and allows for the capture of colonizing tumor cells for a variety of downstream clinical applications.
Collapse
Affiliation(s)
- Brian A Aguado
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Jordan R Caffe
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Dhaval Nanavati
- Proteomics Core Facility, Northwestern University, Chicago, IL 60611, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Grace G Bushnell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
22
|
Dubash AD, Kam CY, Aguado BA, Patel DM, Delmar M, Shea LD, Green KJ. Plakophilin-2 loss promotes TGF-β1/p38 MAPK-dependent fibrotic gene expression in cardiomyocytes. J Cell Biol 2016; 212:425-38. [PMID: 26858265 PMCID: PMC4754716 DOI: 10.1083/jcb.201507018] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/08/2016] [Indexed: 01/07/2023] Open
Abstract
Members of the desmosome protein family are integral components of the cardiac area composita, a mixed junctional complex responsible for electromechanical coupling between cardiomyocytes. In this study, we provide evidence that loss of the desmosomal armadillo protein Plakophilin-2 (PKP2) in cardiomyocytes elevates transforming growth factor β1 (TGF-β1) and p38 mitogen-activated protein kinase (MAPK) signaling, which together coordinate a transcriptional program that results in increased expression of profibrotic genes. Importantly, we demonstrate that expression of Desmoplakin (DP) is lost upon PKP2 knockdown and that restoration of DP expression rescues the activation of this TGF-β1/p38 MAPK transcriptional cascade. Tissues from PKP2 heterozygous and DP conditional knockout mouse models also exhibit elevated TGF-β1/p38 MAPK signaling and induction of fibrotic gene expression in vivo. These data therefore identify PKP2 and DP as central players in coordination of desmosome-dependent TGF-β1/p38 MAPK signaling in cardiomyocytes, pathways known to play a role in different types of cardiac disease, such as arrhythmogenic or hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Adi D Dubash
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 Department of Biology, Furman University, Greenville SC 29613
| | - Chen Y Kam
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Brian A Aguado
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208 Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago IL 60611
| | - Dipal M Patel
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mario Delmar
- New York University School of Medicine, New York, NY 10016
| | - Lonnie D Shea
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208 Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105
| | - Kathleen J Green
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|