1
|
Alshehri AM, Wilson OC. Biomimetic Hydrogel Strategies for Cancer Therapy. Gels 2024; 10:437. [PMID: 39057460 PMCID: PMC11275631 DOI: 10.3390/gels10070437] [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: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Recent developments in biomimetic hydrogel research have expanded the scope of biomedical technologies that can be used to model, diagnose, and treat a wide range of medical conditions. Cancer presents one of the most intractable challenges in this arena due to the surreptitious mechanisms that it employs to evade detection and treatment. In order to address these challenges, biomimetic design principles can be adapted to beat cancer at its own game. Biomimetic design strategies are inspired by natural biological systems and offer promising opportunities for developing life-changing methods to model, detect, diagnose, treat, and cure various types of static and metastatic cancers. In particular, focusing on the cellular and subcellular phenomena that serve as fundamental drivers for the peculiar behavioral traits of cancer can provide rich insights into eradicating cancer in all of its manifestations. This review highlights promising developments in biomimetic nanocomposite hydrogels that contribute to cancer therapies via enhanced drug delivery strategies and modeling cancer mechanobiology phenomena in relation to metastasis and synergistic sensing systems. Creative efforts to amplify biomimetic design research to advance the development of more effective cancer therapies will be discussed in alignment with international collaborative goals to cure cancer.
Collapse
Affiliation(s)
- Awatef M. Alshehri
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
- Department of Nanomedicine, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdelaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia;
| | - Otto C. Wilson
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
2
|
Richbourg NR, Irakoze N, Kim H, Peyton SR. Outlook and opportunities for engineered environments of breast cancer dormancy. SCIENCE ADVANCES 2024; 10:eadl0165. [PMID: 38457510 PMCID: PMC10923521 DOI: 10.1126/sciadv.adl0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/01/2024] [Indexed: 03/10/2024]
Abstract
Dormant, disseminated breast cancer cells resist treatment and may relapse into malignant metastases after decades of quiescence. Identifying how and why these dormant breast cancer cells are triggered into outgrowth is a key unsolved step in treating latent, metastatic breast cancer. However, our understanding of breast cancer dormancy in vivo is limited by technical challenges and ethical concerns with triggering the activation of dormant breast cancer. In vitro models avoid many of these challenges by simulating breast cancer dormancy and activation in well-controlled, bench-top conditions, creating opportunities for fundamental insights into breast cancer biology that complement what can be achieved through animal and clinical studies. In this review, we address clinical and preclinical approaches to treating breast cancer dormancy, how precisely controlled artificial environments reveal key interactions that regulate breast cancer dormancy, and how future generations of biomaterials could answer further questions about breast cancer dormancy.
Collapse
Affiliation(s)
- Nathan R. Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst Amherst, MA 01003, USA
| |
Collapse
|
3
|
Kundu AN, Dougan CE, Mahmoud S, Kilic A, Panagiotou A, Richbourg N, Irakoze N, Peyton SR. Tenascin-C Activation of Lung Fibroblasts in a 3D Synthetic Lung Extracellular Matrix Mimic. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301493. [PMID: 37227134 PMCID: PMC10528529 DOI: 10.1002/adma.202301493] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/26/2023] [Indexed: 05/26/2023]
Abstract
The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that match the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro. Here, a synthetic, bioactive hydrogel is synthesized that mimics the native lung modulus and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin-binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which enables quiescent culture of human lung fibroblasts (HLFs). Stimulation with transforming growth factor β1 (TGF-β1), metastatic breast cancer conditioned media (CM), or tenascin-C-derived integrin-binding peptide activated hydrogel-encapsulated HLFs demonstrates multiple environmental methods to activate HLFs in a lung ECM-mimicking hydrogel. This lung hydrogel platform is a tunable, synthetic approach to studying the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.
Collapse
Affiliation(s)
- Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Carey E. Dougan
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Samar Mahmoud
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Alara Kilic
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst
| | - Alexi Panagiotou
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Nathan Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
- Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003
| |
Collapse
|
4
|
Pasquier E, Rosendahl J, Solberg A, Ståhlberg A, Håkansson J, Chinga-Carrasco G. Polysaccharides and Structural Proteins as Components in Three-Dimensional Scaffolds for Breast Cancer Tissue Models: A Review. Bioengineering (Basel) 2023; 10:682. [PMID: 37370613 PMCID: PMC10295496 DOI: 10.3390/bioengineering10060682] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer is the most common cancer among women, and even though treatments are available, efficiency varies with the patients. In vitro 2D models are commonly used to develop new treatments. However, 2D models overestimate drug efficiency, which increases the failure rate in later phase III clinical trials. New model systems that allow extensive and efficient drug screening are thus required. Three-dimensional printed hydrogels containing active components for cancer cell growth are interesting candidates for the preparation of next generation cancer cell models. Macromolecules, obtained from marine- and land-based resources, can form biopolymers (polysaccharides such as alginate, chitosan, hyaluronic acid, and cellulose) and bioactive components (structural proteins such as collagen, gelatin, and silk fibroin) in hydrogels with adequate physical properties in terms of porosity, rheology, and mechanical strength. Hence, in this study attention is given to biofabrication methods and to the modification with biological macromolecules to become bioactive and, thus, optimize 3D printed structures that better mimic the cancer cell microenvironment. Ink formulations combining polysaccharides for tuning the mechanical properties and bioactive polymers for controlling cell adhesion is key to optimizing the growth of the cancer cells.
Collapse
Affiliation(s)
- Eva Pasquier
- RISE PFI AS, Høgskoleringen 6b, NO-7491 Trondheim, Norway; (E.P.); (A.S.)
| | - Jennifer Rosendahl
- RISE Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, 50115 Borås, Sweden; (J.R.); (J.H.)
| | - Amalie Solberg
- RISE PFI AS, Høgskoleringen 6b, NO-7491 Trondheim, Norway; (E.P.); (A.S.)
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden;
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 41390 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Joakim Håkansson
- RISE Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, 50115 Borås, Sweden; (J.R.); (J.H.)
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, 40530 Gothenburg, Sweden
| | | |
Collapse
|
5
|
Kim H, Wirasaputra A, Mohammadi F, Kundu AN, Esteves JAE, Heiser LM, Meyer AS, Peyton SR. Live Cell Lineage Tracing of Dormant Cancer Cells. Adv Healthc Mater 2023; 12:e2202275. [PMID: 36625629 PMCID: PMC10238615 DOI: 10.1002/adhm.202202275] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/02/2022] [Indexed: 01/11/2023]
Abstract
Breast cancer is a leading cause of global cancer-related deaths, and metastasis is the overwhelming culprit of poor patient prognosis. The most nefarious aspect of metastasis is dormancy, a prolonged period between primary tumor resection and relapse. Current therapies are insufficient at killing dormant cells; thus, they can remain quiescent in the body for decades until eventually undergoing a phenotypic switch, resulting in metastases that are more adaptable and drug resistant. Unfortunately, dormancy has few in vitro models, largely because lab-derived cell lines are highly proliferative. Existing models address tumor dormancy, not cellular dormancy, because tracking individual cells is technically challenging. To combat this problem, a live cell lineage approach to find and track individual dormant cells, distinguishing them from proliferative and dying cells over multiple days, is adapted. This approach is applied across a range of different in vitro microenvironments. This approach reveals that the proportion of cells that exhibit long-term quiescence is regulated by both cell intrinsic and extrinsic factors, with the most dormant cells found in 3D collagen gels. This paper envisions that this approach will prove useful to biologists and bioengineers in the dormancy community to identify, quantify, and study dormant tumor cells.
Collapse
Affiliation(s)
- Hyuna Kim
- Molecular and Cell Biology Graduate Program, University of Massachusetts, Amherst, MA, 01002, USA
| | - Anna Wirasaputra
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Farnaz Mohammadi
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Jennifer A E Esteves
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Shelly R Peyton
- Molecular and Cell Biology Graduate Program, University of Massachusetts, Amherst, MA, 01002, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| |
Collapse
|
6
|
Ahmed T. Functional biomaterials for biomimetic 3D in vitro tumor microenvironment modeling. IN VITRO MODELS 2023; 2:1-23. [PMID: 39872875 PMCID: PMC11756483 DOI: 10.1007/s44164-023-00043-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/30/2025]
Abstract
The translational potential of promising anticancer medications and treatments may be enhanced by the creation of 3D in vitro models that can accurately reproduce native tumor microenvironments. Tumor microenvironments for cancer treatment and research can be built in vitro using biomaterials. Three-dimensional in vitro cancer models have provided new insights into the biology of cancer. Cancer researchers are creating artificial three-dimensional tumor models based on functional biomaterials that mimic the microenvironment of the real tumor. Our understanding of tumor stroma activity over the course of cancer has improved because of the use of scaffold and matrix-based three-dimensional systems intended for regenerative medicine. Scientists have created synthetic tumor models thanks to recent developments in materials engineering. These models enable researchers to investigate the biology of cancer and assess the therapeutic effectiveness of available medications. The emergence of biomaterial engineering technologies with the potential to hasten treatment outcomes is highlighted in this review, which also discusses the influence of creating in vitro biomimetic 3D tumor microenvironments utilizing functional biomaterials. Future cancer treatments will rely much more heavily on biomaterials engineering.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Bashundhara R/A, Dhaka-1229 Dhaka, Bangladesh
| |
Collapse
|
7
|
Peyton SR, Platt MO, Cukierman E. Challenges and Opportunities Modeling the Dynamic Tumor Matrisome. BME FRONTIERS 2023; 4:0006. [PMID: 37849664 PMCID: PMC10521682 DOI: 10.34133/bmef.0006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/28/2022] [Indexed: 10/19/2023] Open
Abstract
We need novel strategies to target the complexity of cancer and, particularly, of metastatic disease. As an example of this complexity, certain tissues are particularly hospitable environments for metastases, whereas others do not contain fertile microenvironments to support cancer cell growth. Continuing evidence that the extracellular matrix (ECM) of tissues is one of a host of factors necessary to support cancer cell growth at both primary and secondary tissue sites is emerging. Research on cancer metastasis has largely been focused on the molecular adaptations of tumor cells in various cytokine and growth factor environments on 2-dimensional tissue culture polystyrene plates. Intravital imaging, conversely, has transformed our ability to watch, in real time, tumor cell invasion, intravasation, extravasation, and growth. Because the interstitial ECM that supports all cells in the tumor microenvironment changes over time scales outside the possible window of typical intravital imaging, bioengineers are continuously developing both simple and sophisticated in vitro controlled environments to study tumor (and other) cell interactions with this matrix. In this perspective, we focus on the cellular unit responsible for upholding the pathologic homeostasis of tumor-bearing organs, cancer-associated fibroblasts (CAFs), and their self-generated ECM. The latter, together with tumoral and other cell secreted factors, constitute the "tumor matrisome". We share the challenges and opportunities for modeling this dynamic CAF/ECM unit, the tools and techniques available, and how the tumor matrisome is remodeled (e.g., via ECM proteases). We posit that increasing information on tumor matrisome dynamics may lead the field to alternative strategies for personalized medicine outside genomics.
Collapse
Affiliation(s)
- Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Manu O. Platt
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| |
Collapse
|
8
|
Creixell M, Kim H, Mohammadi F, Peyton SR, Meyer AS. Systems approaches to uncovering the contribution of environment-mediated drug resistance. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2022; 26:101005. [PMID: 36321161 PMCID: PMC9620953 DOI: 10.1016/j.cossms.2022.101005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cancer drug response is heavily influenced by the extracellular matrix (ECM) environment. Despite a clear appreciation that the ECM influences cancer drug response and progression, a unified view of how, where, and when environment-mediated drug resistance contributes to cancer progression has not coalesced. Here, we survey some specific ways in which the ECM contributes to cancer resistance with a focus on how materials development can coincide with systems biology approaches to better understand and perturb this contribution. We argue that part of the reason that environment-mediated resistance remains a perplexing problem is our lack of a wholistic view of the entire range of environments and their impacts on cell behavior. We cover a series of recent experimental and computational tools that will aid exploration of ECM reactions space, and how they might be synergistically integrated.
Collapse
Affiliation(s)
- Marc Creixell
- Department of Bioengineering, University of California Los Angeles
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Farnaz Mohammadi
- Department of Bioengineering, University of California Los Angeles
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Aaron S Meyer
- Department of Bioengineering, University of California Los Angeles
| |
Collapse
|
9
|
Shah L, Latif A, Williams KJ, Tirella A. Role of stiffness and physico-chemical properties of tumour microenvironment on breast cancer cell stemness. Acta Biomater 2022; 152:273-289. [PMID: 36087866 DOI: 10.1016/j.actbio.2022.08.074] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 01/16/2023]
Abstract
Several physico-chemical properties of the tumour microenvironment (TME) are dysregulated during tumour progression, such as tissue stiffness, extracellular pH and interstitial fluid flow. Traditional preclinical models, although useful to study biological processes, do not provide sufficient control over these physico-chemical properties, hence limiting the understanding of cause-effect relationships between the TME and cancer cells. Breast cancer stem cells (B-CSCs), a dynamic population within the tumour, are known to affect tumour progression, metastasis and therapeutic resistance. With their emerging importance in disease physiology, it is essential to study the interplay between above-mentioned TME physico-chemical variables and B-CSC marker expression. In this work, 3D in vitro models with controlled physico-chemical properties (hydrogel stiffness and composition, perfusion, pH) were used to mimic normal and tumour breast tissue to study changes in proliferation, morphology and B-CSC population in two separate breast cancer cell lines (MCF-7 and MDA-MB 231). Cells encapsulated in alginate-gelatin hydrogels varying in stiffness (2-10 kPa), density and adhesion ligand (gelatin) were perfused (500 µL/min) for up to 14 days. Physiological (pH 7.4) and tumorigenic (pH 6.5) media were used to mimic changes in extracellular pH within the TME. We found that both cell lines have distinct responses to changes in physico-chemical factors in terms of proliferation, cell aggregates size and morphology. Most importantly, stiff and dense hydrogels (10 kPa) and acidic pH (6.5) play a key role in B-CSCs dynamics, increasing both epithelial (E-CSCs) and mesenchymal cancer stem cell (M-CSCs) marker expression, supporting direct impact of the physico-chemical microenvironment on disease onset and progression. STATEMENT OF SIGNIFICANCE: Currently no studies evaluate the impact of physico-chemical properties of the tumour microenvironment on breast cancer stem cell (B-CSC) marker expression in a single in vitro model and at the same time. In this study, 3D in vitro models with varying stiffness, extracellular pH and fluid flow are used to recapitulate the breast tumour microenvironment to evaluate for the first time their direct effect on multiple breast cancer phenotypes: cell proliferation, cell aggregate size and shape, and B-CSC markers. Results suggest these models could open new ways of monitoring disease phenotypes, from the early-onset to progression, as well as being used as testing platforms for effective identification of specific phenotypes in the presence of relevant tumour physico-chemical microenvironment.
Collapse
Affiliation(s)
- Lekha Shah
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, M13 9PL, Manchester, United Kingdom
| | - Ayşe Latif
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, M13 9PL, Manchester, United Kingdom
| | - Kaye J Williams
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, M13 9PL, Manchester, United Kingdom
| | - Annalisa Tirella
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, M13 9PL, Manchester, United Kingdom; BIOtech - Center for Biomedical Technologies, Department of Industrial Engineering, University of Trento, Via delle Regole 101, Trento 38123, Italy.
| |
Collapse
|
10
|
Breast Cancer Patient-Derived Scaffolds Can Expose Unique Individual Cancer Progressing Properties of the Cancer Microenvironment Associated with Clinical Characteristics. Cancers (Basel) 2022; 14:cancers14092172. [PMID: 35565301 PMCID: PMC9103124 DOI: 10.3390/cancers14092172] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Despite huge progress in cancer diagnostics and medicine we still lack optimal cancer treatments for patients with aggressive diseases. This problem can be influenced by the biological heterogeneity of cancer cells as well as poorly understood cancer promoting effects of the cancer microenvironment being an important part of the cancer niche. In this study we have specifically monitored the activity of the cancer microenvironment in breast cancer patients using cell-free scaffolds repopulated with reporter cancer cells sensing the activity of the patient environment. The data show that scaffold induced changes in epithelial-mesenchymal transition and pluripotency markers were linked to clinical and prognostic properties of the original cancer and the information was even more precise when matching estrogen receptor status in our system. The findings highlight that patient-derived scaffolds uncover important information about varying malignant promoting properties in the cancer niche and can be used as a complementary diagnostic tool. Abstract Breast cancer is a heterogeneous disease in terms of cellular and structural composition, and besides acquired aggressive properties in the cancer cell population, the surrounding tumor microenvironment can affect disease progression and clinical behaviours. To specifically decode the clinical relevance of the cancer promoting effects of individual tumor microenvironments, we performed a comprehensive test of 110 breast cancer samples using a recently established in vivo-like 3D cell culture platform based on patient-derived scaffolds (PDSs). Cell-free PDSs were recellularized with three breast cancer cell lines and adaptation to the different patient-based microenvironments was monitored by quantitative PCR. Substantial variability in gene expression between individual PDS cultures from different patients was observed, as well as between different cell lines. Interestingly, specific gene expression changes in the PDS cultures were significantly linked to prognostic features and clinical information from the original cancer. This link was even more pronounced when ERα-status of cell lines and PDSs matched. The results support that PDSs cultures, including a cancer cell line of relevant origin, can monitor the activity of the tumor microenvironment and reveal unique information about the malignancy-inducing properties of the individual cancer niche and serve as a future complementary diagnostic tool for breast cancer.
Collapse
|
11
|
Zhou J, Wu L, Xu P, Li Y, Ji Z, Kang X. Filamin A Is a Potential Driver of Breast Cancer Metastasis via Regulation of MMP-1. Front Oncol 2022; 12:836126. [PMID: 35359350 PMCID: PMC8962737 DOI: 10.3389/fonc.2022.836126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 01/01/2023] Open
Abstract
Recurrent metastasis is a major fatal cause of breast cancer. Regretfully, the driving force and the molecular beneath have not been fully illustrated yet. In this study, a cohort of breast cancer patients with locoregional metastasis was recruited. For them, we collected the matched samples of the primary tumor and metastatic tumor, and then we determined the mutation profiles with whole-exome sequencing (WES). On basis of the profiles, we identified a list of deleterious variants in eight susceptible genes. Of them, filamin A (FLNA) was considered a potential driver gene of metastasis, and its low expression could enhance 5 years’ relapse survival rate by 15%. To prove the finding, we constructed a stable FLNA knockout tumor cell line, which manifested that the cell abilities of proliferation, migration, and invasion were significantly weakened in response to the gene knockout. Subsequently, xenograft mouse experiments further proved that FLNA knockout could inhibit local or distal metastasis. Putting all the results together, we consolidated that FLNA could be a potential driver gene to metastasis of breast cancer, in particular triple-negative breast cancer. Additional experiments also suggested that FLNA might intervene in metastasis via the regulation of MMP-1 expression. In summary, this study demonstrates that FLNA may play as a positive regulator in cancer proliferation and recurrence. It provides new insight into breast cancer metastasis and suggests a potential new therapeutic target for breast cancer therapy.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lvying Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Pengyan Xu
- Department of Surgical Research, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yue Li
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhiliang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- *Correspondence: Xinmei Kang, ; Zhiliang Ji,
| | - Xinmei Kang
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Xinmei Kang, ; Zhiliang Ji,
| |
Collapse
|
12
|
DeCastro AJL, Pranda MA, Gray KM, Merlo-Coyne J, Girma N, Hurwitz M, Zhang Y, Stroka KM. Morphological Phenotyping of Organotropic Brain- and Bone-Seeking Triple Negative Metastatic Breast Tumor Cells. Front Cell Dev Biol 2022; 10:790410. [PMID: 35252171 PMCID: PMC8891987 DOI: 10.3389/fcell.2022.790410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Triple negative breast cancer (TNBC) follows a non-random pattern of metastasis to the bone and brain tissue. Prior work has found that brain-seeking breast tumor cells display altered proteomic profiles, leading to alterations in pathways related to cell signaling, cell cycle, metabolism, and extracellular matrix remodeling. Given the unique microenvironmental characteristics of brain and bone tissue, we hypothesized that brain- or bone-seeking TNBC cells may have altered morphologic or migratory phenotypes from each other, or from the parental TNBC cells, as a function of the biochemical or mechanical microenvironment. In this study, we utilized TNBC cells (MDA-MB-231) that were conditioned to metastasize solely to brain (MDA-BR) or bone (MDA-BO) tissue. We quantified characteristics such as cell morphology, migration, and stiffness in response to cues that partially mimic their final metastatic niche. We have shown that MDA-BO cells have a distinct protrusive morphology not found in MDA-P or MDA-BR. Further, MDA-BO cells migrate over a larger area when on a collagen I (abundant in bone tissue) substrate when compared to fibronectin (abundant in brain tissue). However, migration in highly confined environments was similar across the cell types. Modest differences were found in the stiffness of MDA-BR and MDA-BO cells plated on collagen I vs. fibronectin-coated surfaces. Lastly, MDA-BO cells were found to have larger focal adhesion area and density in comparison with the other two cell types. These results initiate a quantitative profile of mechanobiological phenotypes in TNBC, with future impacts aiming to help predict metastatic propensities to organ-specific sites in a clinical setting.
Collapse
Affiliation(s)
- Ariana Joy L. DeCastro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Marina A. Pranda
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Kelsey M. Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - John Merlo-Coyne
- Department of Biology, University of Maryland, College Park, MD, United States
| | - Nathaniel Girma
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Madelyn Hurwitz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
- Biophysics Program, University of Maryland, College Park, MD, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD, United States
- *Correspondence: Kimberly M. Stroka,
| |
Collapse
|
13
|
Identification of Core Genes and Pathways in Melanoma Metastasis via Bioinformatics Analysis. Int J Mol Sci 2022; 23:ijms23020794. [PMID: 35054979 PMCID: PMC8775799 DOI: 10.3390/ijms23020794] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/05/2023] Open
Abstract
Metastasis is the leading cause of melanoma-related mortality. Current therapies are rarely curative for metastatic melanoma, revealing the urgent need to identify more effective preventive and therapeutic targets. This study aimed to screen the core genes and molecular mechanisms related to melanoma metastasis. A gene expression profile, GSE8401, including 31 primary melanoma and 52 metastatic melanoma clinical samples, was downloaded from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) between melanoma metastases and primary melanoma were screened using GEO2R tool. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) analyses of DEGs were performed using the Database for Annotation Visualization and Integrated Discovery (DAVID). The Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape with Molecular Complex Detection (MCODE) plug-in tools were utilized to detect the protein–protein interaction (PPI) network among DEGs. The top 10 genes with the highest degrees of the PPI network were defined as hub genes. In the results, 425 DEGs, including 60 upregulated genes and 365 downregulated genes, were identified. The upregulated genes were enriched in ECM–receptor interactions and the regulation of actin cytoskeleton, while 365 downregulated genes were enriched in amoebiasis, melanogenesis, and ECM–receptor interactions. The defined hub genes included CDK1, COL17A1, EGFR, DSG1, KRT14, FLG, CDH1, DSP, IVL, and KRT5. In addition, the mRNA and protein levels of the hub genes during melanoma metastasis were verified in the TCGA database and paired post- and premetastatic melanoma cells, respectively. Finally, KRT5-specific siRNAs were utilized to reduce the KRT5 expression in melanoma A375 cells. An MTT assay and a colony formation assay showed that KRT5 knockdown significantly promoted the proliferation of A375 cells. A Transwell assay further suggested that KRT5 knockdown significantly increased the cell migration and cell invasion of A375 cells. This bioinformatics study provided a deeper understanding of the molecular mechanisms of melanoma metastasis. The in vitro experiments showed that KRT5 played the inhibitory effects on melanoma metastasis. Therefore, KRT5 may serve important roles in melanoma metastasis.
Collapse
|
14
|
Jansen LE, Kim H, Hall CL, McCarthy TP, Lee MJ, Peyton SR. A poly(ethylene glycol) three-dimensional bone marrow hydrogel. Biomaterials 2022; 280:121270. [PMID: 34890973 PMCID: PMC8890749 DOI: 10.1016/j.biomaterials.2021.121270] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023]
Abstract
Three-dimensional (3D) hydrogels made from synthetic polymers have emerged as in vitro cell culture platforms capable of representing the extracellular geometry, modulus, and water content of tissues in a tunable fashion. Hydrogels made from these otherwise non-bioactive polymers can be decorated with short peptides derived from proteins naturally found in tissues to support cell viability and direct phenotype. We identified two key limitations that limit the ability of this class of materials to recapitulate real tissue. First, these environments typically display between 1 and 3 bioactive peptides, which vastly underrepresents the diversity of proteins found in the extracellular matrix (ECM) of real tissues. Second, peptides chosen are ubiquitous in ECM and not derived from proteins found in specific tissues, per se. To overcome this critical limitation in hydrogel design and functionality, we developed an approach to incorporate the complex and specific protein signature of bone marrow into a poly (ethylene glycol) (PEG) hydrogel. This bone marrow hydrogel mimics the elasticity of marrow and has 20 bone marrow-specific and cell-instructive peptides. We propose this tissue-centric approach as the next generation of 3D hydrogel design for applications in tissue engineering and beyond.
Collapse
Affiliation(s)
- Lauren E Jansen
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, USA
| | - Christopher L Hall
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Thomas P McCarthy
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Michael J Lee
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, USA; Institute for Applied Life Sciences, University of Massachusetts Amherst 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA, 01003, USA.
| |
Collapse
|
15
|
Tian Z, Wu L, Yu C, Chen Y, Xu Z, Bado I, Loredo A, Wang L, Wang H, Wu KL, Zhang W, Zhang XHF, Xiao H. Harnessing the power of antibodies to fight bone metastasis. SCIENCE ADVANCES 2021; 7:7/26/eabf2051. [PMID: 34162538 PMCID: PMC8221630 DOI: 10.1126/sciadv.abf2051] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/10/2021] [Indexed: 05/04/2023]
Abstract
Antibody-based therapies have proved to be of great value in cancer treatment. Despite the clinical success of these biopharmaceuticals, reaching targets in the bone microenvironment has proved to be difficult due to the relatively low vascularization of bone tissue and the presence of physical barriers. Here, we have used an innovative bone-targeting (BonTarg) technology to generate a first-in-class bone-targeting antibody. Our strategy involves the use of pClick antibody conjugation technology to chemically couple the bone-targeting moiety bisphosphonate to therapeutic antibodies. Bisphosphonate modification of these antibodies results in the delivery of higher conjugate concentrations to the bone metastatic niche, relative to other tissues. In xenograft mice models, this strategy provides enhanced inhibition of bone metastases and multiorgan secondary metastases that arise from bone lesions. Specific delivery of therapeutic antibodies to the bone, therefore, represents a promising strategy for the treatment of bone metastatic cancers and other bone diseases.
Collapse
Affiliation(s)
- Zeru Tian
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Ling Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Chenfei Yu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Yuda Chen
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Zhan Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Igor Bado
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Axel Loredo
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Lushun Wang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Hai Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Kuan-Lin Wu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Weijie Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.
| | - Han Xiao
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, USA.
- Department of Biosciences, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| |
Collapse
|
16
|
Wilson RL, Swaminathan G, Ettayebi K, Bomidi C, Zeng XL, Blutt SE, Estes MK, Grande-Allen KJ. Protein-Functionalized Poly(ethylene glycol) Hydrogels as Scaffolds for Monolayer Organoid Culture. Tissue Eng Part C Methods 2021; 27:12-23. [PMID: 33334213 PMCID: PMC7826425 DOI: 10.1089/ten.tec.2020.0306] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Stem cell-derived, organotypic in vitro models, known as organoids, have emerged as superior alternatives to traditional cell culture models due to their unparalleled ability to recreate complex physiological and pathophysiological processes. For this reason, they are attractive targets of tissue-engineering efforts, as constructs that include organoid technology would be expected to better simulate the many functions of the desired tissue or organ. While the 3D spheroidal architecture that is the default architecture of most organoid models may be preferred for some applications, 2D monolayer arrangements remain the preferred organization for many applications in tissue engineering. Therefore, in this work, we present a method to create monolayer organoid cultures on poly(ethylene glycol) (PEG) hydrogel scaffolds, using intestinal epithelial organoids (IEOs) as a proof-of-concept. Our process involves two steps: the hydrogel is first functionalized with a layer of poly(D-lysine) (PDL), which then allows the adsorption of pristine, unmodified basement membrane proteins. This approach successfully mediates the formation of IEO monolayer unlike conventional approaches that rely on covalent modification of the hydrogel surface with cell-adhesive peptides and basement membrane proteins. We show that these IEO monolayers recreate important physiological functions of the native intestinal epithelium, including multilineage differentiation, apical-basal polarization, and the ability to model infections with human norovirus. We also show coating of a scaffold mimicking intestinal villous topography, resulting in a 3D IEO monolayer. We expect that this protocol will be useful to researchers attempting to leverage the increased physiological relevance of organoid models to elevate the potential of their tissue-engineered constructs. Impact statement While organoids are physiologically superior models of biological functions than traditional cell cultures, their 3D spheroidal architecture is an obstacle to their incorporation in many tissue-engineering applications, which often prefer 2D monolayer arrangements of cells. For this reason, we developed a protocol to establish monolayer cultures of organoids on poly(ethylene glycol) hydrogels and demonstrate its utility using intestinal epithelial organoids as a proof-of-concept. We expect that this protocol will be of use to researchers creating engineered tissues for both regenerative medicine applications, as well as advanced in vitro experimental models.
Collapse
Affiliation(s)
- Reid L. Wilson
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | | | - Khalil Ettayebi
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Carolyn Bomidi
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Xi-Lei Zeng
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah E. Blutt
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K. Estes
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
17
|
Lee J, Abdeen AA, Li Y, Goonetilleke S, Kilian KA. Gradient and Dynamic Hydrogel Materials to Probe Dynamics in Cancer Stem Cell Phenotypes. ACS APPLIED BIO MATERIALS 2020. [DOI: 10.1021/acsabm.0c01263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Junmin Lee
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Amr A. Abdeen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yanfen Li
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Shamalee Goonetilleke
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Kristopher A. Kilian
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- School of Chemistry, School of Materials Science and Engineering, Australian Centre for NanoMedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
18
|
Micek HM, Visetsouk MR, Masters KS, Kreeger PK. Engineering the Extracellular Matrix to Model the Evolving Tumor Microenvironment. iScience 2020; 23:101742. [PMID: 33225247 PMCID: PMC7666341 DOI: 10.1016/j.isci.2020.101742] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical evidence supports a role for the extracellular matrix (ECM) in cancer risk and prognosis across multiple tumor types, and numerous studies have demonstrated that individual ECM components impact key hallmarks of tumor progression (e.g., proliferation, migration, angiogenesis). However, the ECM is a complex network of fibrillar proteins, glycoproteins, and proteoglycans that undergoes dramatic changes in composition and organization during tumor development. In this review, we will highlight how engineering approaches can be used to examine the impact of changes in tissue architecture, ECM composition (i.e., identity and levels of individual ECM components), and cellular- and tissue-level mechanics on tumor progression. In addition, we will discuss recently developed methods to model the ECM that have not yet been applied to the study of cancer.
Collapse
Affiliation(s)
- Hannah M. Micek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mike R. Visetsouk
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kristyn S. Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Pamela K. Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
19
|
Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun 2020; 11:5120. [PMID: 33037194 PMCID: PMC7547708 DOI: 10.1038/s41467-020-18794-x] [Citation(s) in RCA: 1256] [Impact Index Per Article: 251.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Tissues are dynamically shaped by bidirectional communication between resident cells and the extracellular matrix (ECM) through cell-matrix interactions and ECM remodelling. Tumours leverage ECM remodelling to create a microenvironment that promotes tumourigenesis and metastasis. In this review, we focus on how tumour and tumour-associated stromal cells deposit, biochemically and biophysically modify, and degrade tumour-associated ECM. These tumour-driven changes support tumour growth, increase migration of tumour cells, and remodel the ECM in distant organs to allow for metastatic progression. A better understanding of the underlying mechanisms of tumourigenic ECM remodelling is crucial for developing therapeutic treatments for patients. Tumors are more than cancer cells — the extracellular matrix is a protein structure that organizes all tissues and is altered in cancer. Here, the authors review recent progress in understanding how the cancer cells and tumor-associated stroma cells remodel the extracellular matrix to drive tumor growth and metastasis.
Collapse
Affiliation(s)
- Juliane Winkler
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA.
| | - Abisola Abisoye-Ogunniyan
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Kevin J Metcalf
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Zena Werb
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
20
|
Valcourt DM, Kapadia CH, Scully MA, Dang MN, Day ES. Best Practices for Preclinical In Vivo Testing of Cancer Nanomedicines. Adv Healthc Mater 2020; 9:e2000110. [PMID: 32367687 PMCID: PMC7473451 DOI: 10.1002/adhm.202000110] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/17/2020] [Indexed: 01/06/2023]
Abstract
Significant advances have been made in the development of nanoparticles for cancer treatment in recent years. Despite promising results in preclinical animal models, cancer nanomedicines often fail in clinical trials. This failure rate could be reduced by defining stringent criteria for testing and quality control during the design and development stages, and by performing carefully planned preclinical studies in relevant animal models. This article discusses best practices for the evaluation of nanomedicines in murine tumor models. First, a recommended set of experiments to perform is introduced, including discussion of the types of data to collect during these studies. This is followed by an outline of various tumor models and their clinical relevance. Next, different routes of nanoparticle administration are overviewed, followed by a summary of important controls to include in in vivo studies of nanomedicine. Finally, animal welfare considerations are discussed, and an overview of the steps involved in achieving US Food and Drug Administration approval after animal studies are completed is provided. Researchers should use this report as a guideline for effective preclinical evaluation of cancer nanomedicine. As the community adopts best practices for in vivo testing, the rate of clinical translation of cancer nanomedicines is likely to improve.
Collapse
Affiliation(s)
- Danielle M Valcourt
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Chintan H Kapadia
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Mackenzie A Scully
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Megan N Dang
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science & Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
- Helen F. Graham Cancer Center & Research Institute, 4701 Ogletown Stanton Road, Newark, DE, 19713, USA
| |
Collapse
|
21
|
Bittner KR, Jiménez JM, Peyton SR. Vascularized Biomaterials to Study Cancer Metastasis. Adv Healthc Mater 2020; 9:e1901459. [PMID: 31977160 PMCID: PMC7899188 DOI: 10.1002/adhm.201901459] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/07/2019] [Indexed: 12/15/2022]
Abstract
Cancer metastasis, the spread of cancer cells to distant organs, is responsible for 90% of cancer-related deaths. Cancer cells need to enter and exit circulation in order to form metastases, and the vasculature and endothelial cells are key regulators of this process. While vascularized 3D in vitro systems have been developed, few have been used to study cancer, and many lack key features of vessels that are necessary to study metastasis. This review focuses on current methods of vascularizing biomaterials for the study of cancer, and three main factors that regulate intravasation and extravasation: endothelial cell heterogeneity, hemodynamics, and the extracellular matrix of the perivascular niche.
Collapse
Affiliation(s)
- Katharine R Bittner
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
| | - Juan M Jiménez
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
22
|
Barney LE, Hall CL, Schwartz AD, Parks AN, Sparages C, Galarza S, Platt MO, Mercurio AM, Peyton SR. Tumor cell-organized fibronectin maintenance of a dormant breast cancer population. SCIENCE ADVANCES 2020; 6:eaaz4157. [PMID: 32195352 PMCID: PMC7065904 DOI: 10.1126/sciadv.aaz4157] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/17/2019] [Indexed: 05/04/2023]
Abstract
Tumors can undergo long periods of dormancy, with cancer cells entering a largely quiescent, nonproliferative state before reactivation and outgrowth. To understand the role of the extracellular matrix (ECM) in regulating tumor dormancy, we created an in vitro cell culture system with carefully controlled ECM substrates to observe entrance into and exit from dormancy with live imaging. We saw that cell populations capable of surviving entrance into long-term dormancy were heterogeneous, containing quiescent, cell cycle-arrested, and actively proliferating cells. Cell populations capable of entering dormancy formed an organized, fibrillar fibronectin matrix via αvβ3 and α5β1 integrin adhesion, ROCK-generated tension, and TGFβ2 stimulation, and cancer cell outgrowth after dormancy required MMP-2-mediated fibronectin degradation. We propose this approach as a useful, in vitro method to study factors important in regulating dormancy, and we used it here to elucidate a role for fibronectin deposition and MMP activation.
Collapse
Affiliation(s)
- Lauren E. Barney
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Christopher L. Hall
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Alyssa D. Schwartz
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Akia N. Parks
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332, USA
| | - Christopher Sparages
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Sualyneth Galarza
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Manu O. Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332, USA
| | - Arthur M. Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| |
Collapse
|
23
|
Galarza S, Crosby AJ, Pak C, Peyton SR. Control of Astrocyte Quiescence and Activation in a Synthetic Brain Hydrogel. Adv Healthc Mater 2020; 9:e1901419. [PMID: 31943839 DOI: 10.1002/adhm.201901419] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Bioengineers have designed numerous instructive brain extracellular matrix (ECM) environments with tailored and tunable protein compositions and biomechanical properties in vitro to study astrocyte reactivity during trauma and inflammation. However, a major limitation of both protein-based and synthetic model microenvironments is that astrocytes within fail to retain their characteristic stellate morphology and quiescent state without becoming activated under "normal" culture conditions. Here, a synthetic hydrogel is introduced, which for the first time demonstrates maintenance of astrocyte quiescence and activation on demand. With this synthetic brain hydrogel, the brain-specific integrin-binding and matrix metalloprotease-degradable domains of proteins are shown to control astrocyte star-shaped morphologies, and an ECM condition that maintains astrocyte quiescence with minimal activation can be achieved. In addition, activation can be induced in a dose-dependent manner via both defined cytokine cocktails and low molecular weight hyaluronic acid. This synthetic brain hydrogel is envisioned as a new tool to study the physiological role of astrocytes in health and disease.
Collapse
Affiliation(s)
- Sualyneth Galarza
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Alfred J. Crosby
- Department of Polymer Science and Engineering University of Massachusetts Amherst MA 01003 USA
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology University of Massachusetts Amherst MA 01003 USA
| | - Shelly R. Peyton
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
24
|
Galarza S, Kim H, Atay N, Peyton SR, Munson JM. 2D or 3D? How cell motility measurements are conserved across dimensions in vitro and translate in vivo. Bioeng Transl Med 2020; 5:e10148. [PMID: 31989037 PMCID: PMC6971446 DOI: 10.1002/btm2.10148] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 12/16/2022] Open
Abstract
Cell motility is a critical aspect of several processes, such as wound healing and immunity; however, it is dysregulated in cancer. Current limitations of imaging tools make it difficult to study cell migration in vivo. To overcome this, and to identify drivers from the microenvironment that regulate cell migration, bioengineers have developed 2D (two-dimensional) and 3D (three-dimensional) tissue model systems in which to study cell motility in vitro, with the aim of mimicking elements of the environments in which cells move in vivo. However, there has been no systematic study to explicitly relate and compare cell motility measurements between these geometries or systems. Here, we provide such analysis on our own data, as well as across data in existing literature to understand whether, and which, metrics are conserved across systems. To our surprise, only one metric of cell movement on 2D surfaces significantly and positively correlates with cell migration in 3D environments (percent migrating cells), and cell invasion in 3D has a weak, negative correlation with glioblastoma invasion in vivo. Finally, to compare across complex model systems, in vivo data, and data from different labs, we suggest that groups report an effect size, a statistical tool that is most translatable across experiments and labs, when conducting experiments that affect cellular motility.
Collapse
Affiliation(s)
- Sualyneth Galarza
- Department of Chemical EngineeringUniversity of Massachusetts AmherstAmherstMassachusetts
| | - Hyuna Kim
- Molecular and Cellular Biology ProgramUniversity of Massachusetts AmherstAmherstMassachusetts
| | - Naciye Atay
- Department of Biomedical Engineering and MechanicsVirginia Polytechnic Institute and State UniversityBlacksburgVirginia
| | - Shelly R. Peyton
- Department of Chemical EngineeringUniversity of Massachusetts AmherstAmherstMassachusetts
- Molecular and Cellular Biology ProgramUniversity of Massachusetts AmherstAmherstMassachusetts
| | - Jennifer M. Munson
- Department of Biomedical Engineering and MechanicsVirginia Polytechnic Institute and State UniversityBlacksburgVirginia
| |
Collapse
|
25
|
Schutrum BE, Whitman MA, Fischbach C. Biomaterials-Based Model Systems to Study Tumor–Microenvironment Interactions. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00077-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
Bray LJ, Hutmacher DW, Bock N. Addressing Patient Specificity in the Engineering of Tumor Models. Front Bioeng Biotechnol 2019; 7:217. [PMID: 31572718 PMCID: PMC6751285 DOI: 10.3389/fbioe.2019.00217] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer treatment is challenged by the heterogeneous nature of cancer, where prognosis depends on tumor type and disease stage, as well as previous treatments. Optimal patient stratification is critical for the development and validation of effective treatments, yet pre-clinical model systems are lacking in the delivery of effective individualized platforms that reflect distinct patient-specific clinical situations. Advances in cancer cell biology, biofabrication, and microengineering technologies have led to the development of more complex in vitro three-dimensional (3D) models to act as drug testing platforms and to elucidate novel cancer mechanisms. Mostly, these strategies have enabled researchers to account for the tumor microenvironment context including tumor-stroma interactions, a key factor of heterogeneity that affects both progression and therapeutic resistance. This is aided by state-of-the-art biomaterials and tissue engineering technologies, coupled with reproducible and high-throughput platforms that enable modeling of relevant physical and chemical factors. Yet, the translation of these models and technologies has been impaired by neglecting to incorporate patient-derived cells or tissues, and largely focusing on immortalized cell lines instead, contributing to drug failure rates. While this is a necessary step to establish and validate new models, a paradigm shift is needed to enable the systematic inclusion of patient-derived materials in the design and use of such models. In this review, we first present an overview of the components responsible for heterogeneity in different tumor microenvironments. Next, we introduce the state-of-the-art of current in vitro 3D cancer models employing patient-derived materials in traditional scaffold-free approaches, followed by novel bioengineered scaffold-based approaches, and further supported by dynamic systems such as bioreactors, microfluidics, and tumor-on-a-chip devices. We critically discuss the challenges and clinical prospects of models that have succeeded in providing clinical relevance and impact, and present emerging concepts of novel cancer model systems that are addressing patient specificity, the next frontier to be tackled by the field.
Collapse
Affiliation(s)
- Laura J. Bray
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Dietmar W. Hutmacher
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
- Australian Research Council (ARC) Industrial Transformation Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
- Translational Research Institute, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane, QLD, Australia
| |
Collapse
|
27
|
Abstract
Cell adhesion to the extracellular matrix is fundamental to tissue integrity and human health. Integrins are the main cellular adhesion receptors that through multifaceted roles as signalling molecules, mechanotransducers and key components of the cell migration machinery are implicated in nearly every step of cancer progression from primary tumour development to metastasis. Altered integrin expression is frequently detected in tumours, where integrins have roles in supporting oncogenic growth factor receptor (GFR) signalling and GFR-dependent cancer cell migration and invasion. In addition, integrins determine colonization of metastatic sites and facilitate anchorage-independent survival of circulating tumour cells. Investigations describing integrin engagement with a growing number of versatile cell surface molecules, including channels, receptors and secreted proteins, continue to lead to the identification of novel tumour-promoting pathways. Integrin-mediated sensing, stiffening and remodelling of the tumour stroma are key steps in cancer progression supporting invasion, acquisition of cancer stem cell characteristics and drug resistance. Given the complexity of integrins and their adaptable and sometimes antagonistic roles in cancer cells and the tumour microenvironment, therapeutic targeting of these receptors has been a challenge. However, novel approaches to target integrins and antagonism of specific integrin subunits in stringently stratified patient cohorts are emerging as potential ways forward.
Collapse
Affiliation(s)
- Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Biochemistry, University of Turku, Turku, Finland.
| |
Collapse
|
28
|
Schwartz AD, Hall CL, Barney LE, Babbitt CC, Peyton SR. Integrin α 6 and EGFR signaling converge at mechanosensitive calpain 2. Biomaterials 2018; 178:73-82. [PMID: 29909039 PMCID: PMC6211197 DOI: 10.1016/j.biomaterials.2018.05.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/14/2018] [Accepted: 05/31/2018] [Indexed: 11/30/2022]
Abstract
Cells sense and respond to mechanical cues from the extracellular matrix (ECM) via integrins. ECM stiffness is known to enhance integrin clustering and response to epidermal growth factor (EGF), but we lack information on when or if these mechanosensitive growth factor receptors and integrins converge intracellularly. Towards closing this knowledge gap, we combined a biomaterial platform with transcriptomics, molecular biology, and functional assays to link integrin-mediated mechanosensing and epidermal growth factor receptor (EGFR) signaling. We found that high integrin α6 expression controlled breast cancer cell adhesion and motility on soft, laminin-coated substrates, and this mimicked the response of cells to EGF stimulation. The mechanisms that drove both mechanosensitive cell adhesion and motility converged on calpain 2, an intracellular protease important for talin cleavage and focal adhesion turnover. EGF stimulation enhanced adhesion and motility on soft substrates, but required integrin α6 and calpain 2 signaling. In sum, we identified a new role for integrin α6 mechanosensing in breast cancer, wherein cell adhesion to laminin on soft substrates mimicked EGF stimulation. We identified calpain 2, downstream of both integrin α6 engagement and EGFR phosphorylation, as a common intracellular signaling node, and implicate integrin α6 and calpain 2 as potential targets to inhibit the migration of cancer cells in stiff tumor environments.
Collapse
Affiliation(s)
- A D Schwartz
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - C L Hall
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - L E Barney
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - C C Babbitt
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - S R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
29
|
Plou J, Juste-Lanas Y, Olivares V, Del Amo C, Borau C, García-Aznar JM. From individual to collective 3D cancer dissemination: roles of collagen concentration and TGF-β. Sci Rep 2018; 8:12723. [PMID: 30143683 PMCID: PMC6109049 DOI: 10.1038/s41598-018-30683-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer cells have the ability to migrate from the primary (original) site to other places in the body. The extracellular matrix affects cancer cell migratory capacity and has been correlated with tissue-specific spreading patterns. However, how the matrix orchestrates these behaviors remains unclear. Here, we investigated how both higher collagen concentrations and TGF-β regulate the formation of H1299 cell (a non-small cell lung cancer cell line) spheroids within 3D collagen-based matrices and promote cancer cell invasive capacity. We show that at low collagen concentrations, tumor cells move individually and have moderate invasive capacity, whereas when the collagen concentration is increased, the formation of cell clusters is promoted. In addition, when the concentration of TGF-β in the microenvironment is lower, most of the clusters are aggregates of cancer cells with a spheroid-like morphology and poor migratory capacity. In contrast, higher concentrations of TGF-β induced the formation of clusters with a notably higher invasive capacity, resulting in clear strand-like collective cell migration. Our results show that the concentration of the extracellular matrix is a key regulator of the formation of tumor clusters that affects their development and growth. In addition, chemical factors create a microenvironment that promotes the transformation of idle tumor clusters into very active, invasive tumor structures. These results collectively demonstrate the relevant regulatory role of the mechano-chemical microenvironment in leading the preferential metastasis of tumor cells to specific tissues with high collagen concentrations and TFG-β activity.
Collapse
Affiliation(s)
- J Plou
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain.
| | - Y Juste-Lanas
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - V Olivares
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - C Del Amo
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - C Borau
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - J M García-Aznar
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain.
| |
Collapse
|
30
|
Luzhansky ID, Schwartz AD, Cohen JD, MacMunn JP, Barney LE, Jansen LE, Peyton SR. Anomalously diffusing and persistently migrating cells in 2D and 3D culture environments. APL Bioeng 2018; 2:026112. [PMID: 31069309 PMCID: PMC6324209 DOI: 10.1063/1.5019196] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 05/28/2018] [Indexed: 12/17/2022] Open
Abstract
Appropriately chosen descriptive models of cell migration in biomaterials will allow researchers to characterize and ultimately predict the movement of cells in engineered systems for a variety of applications in tissue engineering. The persistent random walk (PRW) model accurately describes cell migration on two-dimensional (2D) substrates. However, this model inherently cannot describe subdiffusive cell movement, i.e., migration paths in which the root mean square displacement increases more slowly than the square root of the time interval. Subdiffusivity is a common characteristic of cells moving in confined environments, such as three-dimensional (3D) porous scaffolds, hydrogel networks, and in vivo tissues. We demonstrate that a generalized anomalous diffusion (AD) model, which uses a simple power law to relate the mean square displacement to time, more accurately captures individual cell migration paths across a range of engineered 2D and 3D environments than does the more commonly used PRW model. We used the AD model parameters to distinguish cell movement profiles on substrates with different chemokinetic factors, geometries (2D vs 3D), substrate adhesivities, and compliances. Although the two models performed with equal precision for superdiffusive cells, we suggest a simple AD model, in lieu of PRW, to describe cell trajectories in populations with a significant subdiffusive fraction, such as cells in confined, 3D environments.
Collapse
Affiliation(s)
- Igor D. Luzhansky
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | - Alyssa D. Schwartz
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | - Joshua D. Cohen
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - John P. MacMunn
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | - Lauren E. Barney
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | - Lauren E. Jansen
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, USA
| |
Collapse
|
31
|
Lee JV, Berry CT, Kim K, Sen P, Kim T, Carrer A, Trefely S, Zhao S, Fernandez S, Barney LE, Schwartz AD, Peyton SR, Snyder NW, Berger SL, Freedman BD, Wellen KE. Acetyl-CoA promotes glioblastoma cell adhesion and migration through Ca 2+-NFAT signaling. Genes Dev 2018; 32:497-511. [PMID: 29674394 PMCID: PMC5959234 DOI: 10.1101/gad.311027.117] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/26/2018] [Indexed: 01/05/2023]
Abstract
Here, Lee et al. investigated the molecular mechanisms by which acetyl-CoA production impacts gene expression and how acetyl-CoA promotes malignant phenotypes. Their findings show that acetyl-CoA can enhance H3K27ac in a locus-specific manner and that expression of cell adhesion genes is driven by acetyl-CoA in part through activation of Ca2+–NFAT signaling. The metabolite acetyl-coenzyme A (acetyl-CoA) is the required acetyl donor for lysine acetylation and thereby links metabolism, signaling, and epigenetics. Nutrient availability alters acetyl-CoA levels in cancer cells, correlating with changes in global histone acetylation and gene expression. However, the specific molecular mechanisms through which acetyl-CoA production impacts gene expression and its functional roles in promoting malignant phenotypes are poorly understood. Here, using histone H3 Lys27 acetylation (H3K27ac) ChIP-seq (chromatin immunoprecipitation [ChIP] coupled with next-generation sequencing) with normalization to an exogenous reference genome (ChIP-Rx), we found that changes in acetyl-CoA abundance trigger site-specific regulation of H3K27ac, correlating with gene expression as opposed to uniformly modulating this mark at all genes. Genes involved in integrin signaling and cell adhesion were identified as acetyl-CoA-responsive in glioblastoma cells, and we demonstrate that ATP citrate lyase (ACLY)-dependent acetyl-CoA production promotes cell migration and adhesion to the extracellular matrix. Mechanistically, the transcription factor NFAT1 (nuclear factor of activated T cells 1) was found to mediate acetyl-CoA-dependent gene regulation and cell adhesion. This occurs through modulation of Ca2+ signals, triggering NFAT1 nuclear translocation when acetyl-CoA is abundant. The findings of this study thus establish that acetyl-CoA impacts H3K27ac at specific loci, correlating with gene expression, and that expression of cell adhesion genes are driven by acetyl-CoA in part through activation of Ca2+–NFAT signaling.
Collapse
Affiliation(s)
- Joyce V Lee
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Corbett T Berry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,School of Biomedical Engineering, Drexel University, Philadelphia, Pennsylvania 19104, USA
| | - Karla Kim
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Payel Sen
- Penn Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Taehyong Kim
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Alessandro Carrer
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,A.J. Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania 19104, USA
| | - Steven Zhao
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Sully Fernandez
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Lauren E Barney
- Department of Chemical Engineering, University of Massachusetts-Amherst, Amherst, Massachusetts 01003, USA
| | - Alyssa D Schwartz
- Department of Chemical Engineering, University of Massachusetts-Amherst, Amherst, Massachusetts 01003, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts-Amherst, Amherst, Massachusetts 01003, USA
| | - Nathaniel W Snyder
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania 19104, USA
| | - Shelley L Berger
- Penn Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA.,Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
32
|
Lourenço BN, Springer NL, Ferreira D, Oliveira C, Granja PL, Fischbach C. CD44v6 increases gastric cancer malignant phenotype by modulating adipose stromal cell-mediated ECM remodeling. Integr Biol (Camb) 2018; 10:145-158. [PMID: 29450424 PMCID: PMC5988203 DOI: 10.1039/c7ib00179g] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CD44, an abundantly expressed adhesion molecule, and its alternative splice variants have been associated with tumorigenesis and metastasis. In the context of gastric cancer (GC), de novo expression of CD44 variant 6 (CD44v6) is found in more than 60% of GCs, but its role in the pathogenesis and progression of this type of cancer remains unclear. Using a combination of media conditioning experiments and decellularized extracellular matrices (ECMs), this study investigates the hypothesis that CD44v6 overexpression enhances tumor cell malignant behavior by modulating stromal cell-mediated ECM remodeling. Our findings indicate that soluble factors secreted by CD44v6 expressing GC cells particularly increase proliferation and myofibroblastic differentiation of adipose stromal cells (ASCs). These changes in ASC phenotype mediate the deposition of fibrotic/desmoplastic ECM that, in turn, stimulates GC proliferation and inhibits GC clustering. Pharmacological inhibition of matrix metalloproteinase (MMP) activity in tumor cells abrogated matrix-induced changes in tumor cell malignant behavior. Additionally, studies in mice confirmed the pathological relevance of CD44v6 expression and consequential changes in ECM remodeling to gastric tumorigenesis in vivo. Collectively, these results indicate a direct link between CD44v6, ECM remodeling, and GC malignant behavior opening new insights into potential CD44v6-targeted therapies.
Collapse
Affiliation(s)
- Bianca N Lourenço
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 157 Weill Hall, Ithaca, NY 14853, USA. and i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal and IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Portugal and Faculdade de Engenharia, Universidade do Porto, Portugal
| | - Nora L Springer
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 157 Weill Hall, Ithaca, NY 14853, USA. and Biological and Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | - Daniel Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal and IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Portugal and Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Portugal and Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Portugal
| | - Pedro L Granja
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal and Faculdade de Engenharia, Universidade do Porto, Portugal and Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 157 Weill Hall, Ithaca, NY 14853, USA. and Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA
| |
Collapse
|
33
|
Brooks EA, Jansen LE, Gencoglu MF, Yurkevicz AM, Peyton SR. Complementary, Semiautomated Methods for Creating Multidimensional PEG-Based Biomaterials. ACS Biomater Sci Eng 2018; 4:707-718. [PMID: 33418758 DOI: 10.1021/acsbiomaterials.7b00737] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tunable biomaterials that mimic selected features of the extracellular matrix (ECM) such as its stiffness, protein composition, and dimensionality are increasingly popular for studying how cells sense and respond to ECM cues. In the field, there exists a significant trade-off for how complex and how well these biomaterials represent the in vivo microenvironment versus how easy they are to make and how adaptable they are to automated fabrication techniques. To address this need to integrate more complex biomaterials design with high-throughput screening approaches, we present several methods to fabricate synthetic biomaterials in 96-well plates and demonstrate that they can be adapted to semiautomated liquid handling robotics. These platforms include (1) glass bottom plates with covalently attached ECM proteins and (2) hydrogels with tunable stiffness and protein composition with either cells seeded on the surface or (3) laden within the three-dimensional hydrogel matrix. This study includes proof-of-concept results demonstrating control over breast cancer cell line phenotypes via these ECM cues in a semiautomated fashion. We foresee the use of these methods as a mechanism to bridge the gap between high-throughput cell-matrix screening and engineered ECM-mimicking biomaterials.
Collapse
Affiliation(s)
- Elizabeth A Brooks
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Lauren E Jansen
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Maria F Gencoglu
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Annali M Yurkevicz
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| |
Collapse
|
34
|
Kittur H, Tay A, Hua A, Yu M, Di Carlo D. Probing Cell Adhesion Profiles with a Microscale Adhesive Choice Assay. Biophys J 2017; 113:1858-1867. [PMID: 29045879 PMCID: PMC5647542 DOI: 10.1016/j.bpj.2017.08.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/13/2017] [Accepted: 08/15/2017] [Indexed: 10/18/2022] Open
Abstract
In this work, we introduce, to our knowledge, a new set of adhesion-based biomarkers for characterizing mammalian cells. Mammalian cell adhesion to the extracellular matrix influences numerous physiological processes. Current in vitro methods to probe adhesion focus on adhesive force to a single surface, which can investigate only a subcomponent of the adhesive, motility, and polarization cues responsible for adhesion in the 3D tissue environment. Here, we demonstrate a method to quantify the transhesive properties of cells that relies on the microscale juxtaposition of two extracellular matrix-coated surfaces. By multiplexing this approach, we investigate the unique transhesive profiles for breast cancer cells that are adapted to colonize different metastatic sites. We find that malignant breast cancer cells readily transfer to new collagen I surfaces, and away from basement membrane proteins. Integrins and actin polymerization largely regulate this transfer. This tool can be readily adopted in cell biology and cancer research to uncover, to our knowledge, novel drivers of adhesion (or de-adhesion) and sort cell populations based on complex phenotypes with physiological relevance.
Collapse
Affiliation(s)
- Harsha Kittur
- University of California Los Angeles, Los Angeles, California
| | - Andy Tay
- University of California Los Angeles, Los Angeles, California
| | - Avery Hua
- University of California Los Angeles, Los Angeles, California
| | - Min Yu
- University of Southern California, Los Angeles, California
| | - Dino Di Carlo
- University of California Los Angeles, Los Angeles, California; California NanoSystems Institute, Los Angeles, California; Jonsson Comprehensive Cancer Center, Los Angeles, California.
| |
Collapse
|
35
|
Multiscale characterization of the mineral phase at skeletal sites of breast cancer metastasis. Proc Natl Acad Sci U S A 2017; 114:10542-10547. [PMID: 28923958 DOI: 10.1073/pnas.1708161114] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Skeletal metastases, the leading cause of death in advanced breast cancer patients, depend on tumor cell interactions with the mineralized bone extracellular matrix. Bone mineral is largely composed of hydroxyapatite (HA) nanocrystals with physicochemical properties that vary significantly by anatomical location, age, and pathology. However, it remains unclear whether bone regions typically targeted by metastatic breast cancer feature distinct HA materials properties. Here we combined high-resolution X-ray scattering analysis with large-area Raman imaging, backscattered electron microscopy, histopathology, and microcomputed tomography to characterize HA in mouse models of advanced breast cancer in relevant skeletal locations. The proximal tibial metaphysis served as a common metastatic site in our studies; we identified that in disease-free bones this skeletal region contained smaller and less-oriented HA nanocrystals relative to ones that constitute the diaphysis. We further observed that osteolytic bone metastasis led to a decrease in HA nanocrystal size and perfection in remnant metaphyseal trabecular bone. Interestingly, in a model of localized breast cancer, metaphyseal HA nanocrystals were also smaller and less perfect than in corresponding bone in disease-free controls. Collectively, these results suggest that skeletal sites prone to tumor cell dissemination contain less-mature HA (i.e., smaller, less-perfect, and less-oriented crystals) and that primary tumors can further increase HA immaturity even before secondary tumor formation, mimicking alterations present during tibial metastasis. Engineered tumor models recapitulating these spatiotemporal dynamics will permit assessing the functional relevance of the detected changes to the progression and treatment of breast cancer bone metastasis.
Collapse
|
36
|
Advanced biomaterials and microengineering technologies to recapitulate the stepwise process of cancer metastasis. Biomaterials 2017; 133:176-207. [DOI: 10.1016/j.biomaterials.2017.04.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/04/2017] [Accepted: 04/12/2017] [Indexed: 02/08/2023]
|
37
|
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
|
38
|
Zhang D, Lee J, Sun MB, Pei Y, Chu J, Gillette MU, Fan TM, Kilian KA. Combinatorial Discovery of Defined Substrates That Promote a Stem Cell State in Malignant Melanoma. ACS CENTRAL SCIENCE 2017; 3:381-393. [PMID: 28573199 PMCID: PMC5445527 DOI: 10.1021/acscentsci.6b00329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Indexed: 06/07/2023]
Abstract
The tumor microenvironment is implicated in orchestrating cancer cell transformation and metastasis. However, specific cell-ligand interactions between cancer cells and the extracellular matrix are difficult to decipher due to a dynamic and multivariate presentation of many signaling molecules. Here we report a versatile peptide microarray platform that is capable of screening for cancer cell phenotypic changes in response to ligand-receptor interactions. Using a screen of 78 peptide combinations derived from proteins present in the melanoma microenvironment, we identify a proteoglycan binding and bone morphogenic protein 7 (BMP7) derived sequence that selectively promotes the expression of several putative melanoma initiating cell markers. We characterize signaling associated with each of these peptides in the activation of melanoma pro-tumorigenic signaling and reveal a role for proteoglycan mediated adhesion and signaling through Smad 2/3. A defined substratum that controls the state of malignant melanoma may prove useful in spatially normalizing a heterogeneous population of tumor cells for discovery of therapeutics that target a specific state and for identifying new drug targets and reagents for intervention.
Collapse
Affiliation(s)
- Douglas Zhang
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Junmin Lee
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Michael B. Sun
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Yi Pei
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - James Chu
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Martha U. Gillette
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Kristopher A. Kilian
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
39
|
Archer TC, Fertig EJ, Gosline SJC, Hafner M, Hughes SK, Joughin BA, Meyer AS, Piccolo SR, Shajahan-Haq AN. Systems Approaches to Cancer Biology. Cancer Res 2016; 76:6774-6777. [PMID: 27864348 PMCID: PMC5135591 DOI: 10.1158/0008-5472.can-16-1580] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/27/2016] [Accepted: 09/12/2016] [Indexed: 01/30/2023]
Abstract
Cancer systems biology aims to understand cancer as an integrated system of genes, proteins, networks, and interactions rather than an entity of isolated molecular and cellular components. The inaugural Systems Approaches to Cancer Biology Conference, cosponsored by the Association of Early Career Cancer Systems Biologists and the National Cancer Institute of the NIH, focused on the interdisciplinary field of cancer systems biology and the challenging cancer questions that are best addressed through the combination of experimental and computational analyses. Attendees found that elucidating the many molecular features of cancer inevitably reveals new forms of complexity and concluded that ensuring the reproducibility and impact of cancer systems biology studies will require widespread method and data sharing and, ultimately, the translation of important findings to the clinic. Cancer Res; 76(23); 6774-7. ©2016 AACR.
Collapse
Affiliation(s)
- Tenley C Archer
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts
| | - Elana J Fertig
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | | | - Marc Hafner
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| | - Shannon K Hughes
- Division of Cancer Biology, National Cancer Institute of the NIH, Rockville, Maryland
| | - Brian A Joughin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Aaron S Meyer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| | | | - Ayesha N Shajahan-Haq
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
40
|
Maji S, Chaudhary P, Akopova I, Nguyen PM, Hare RJ, Gryczynski I, Vishwanatha JK. Exosomal Annexin II Promotes Angiogenesis and Breast Cancer Metastasis. Mol Cancer Res 2016; 15:93-105. [PMID: 27760843 DOI: 10.1158/1541-7786.mcr-16-0163] [Citation(s) in RCA: 252] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/05/2016] [Accepted: 10/03/2016] [Indexed: 12/18/2022]
Abstract
Tumor-derived exosomes are emerging mediators of tumorigenesis and tissue-specific metastasis. Proteomic profiling has identified Annexin II as one of the most highly expressed proteins in exosomes; however, studies focused on the biological role of exosomal Annexin II (exo-Anx II) are still lacking. In this study, mechanistic insight was sought regarding exo-Anx II and its function in angiogenesis and breast cancer metastasis. Multiple in vitro and in vivo techniques were used to study the role of exo-Anx II in angiogenesis. Using atomic force microscopy and Western blotting, exo-Anx II expression was characterized in normal and breast cancer cells. In addition, organ-specific metastatic breast cancer cells and animal models were used to define the role exo-Anx II in breast cancer metastasis. Results revealed that exo-Anx II expression is significantly higher in malignant cells than normal and premetastatic breast cancer cells. In vitro and in vivo studies demonstrated that exo-Anx II promotes tPA-dependent angiogenesis. Furthermore, in vivo analysis indicated that metastatic exosomes create a favorable microenvironment for metastasis, and exo-Anx II plays an important role in this process, as priming with Anx II-depleted exosomes reduces brain (∼4-fold) and lung (∼2-fold) metastasis. Upon delineating the mechanism, it was discovered that exo-Anx II causes macrophage-mediated activation of the p38MAPK, NF-κB, and STAT3 pathways and increased secretion of IL6 and TNFα. These data demonstrate an important role for exo-Anx II in breast cancer pathogenesis. IMPLICATIONS Exosome-associated Annexin II plays an important role in angiogenesis and breast cancer metastasis, which can be exploited as a potential biomarker as well as a therapeutic target for diagnosis and treatment of metastatic breast cancer. Mol Cancer Res; 15(1); 93-105. ©2016 AACR.
Collapse
Affiliation(s)
- Sayantan Maji
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas. .,Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas
| | - Pankaj Chaudhary
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas.,Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas
| | - Irina Akopova
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | - Phung M Nguyen
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | | | - Ignacy Gryczynski
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | - Jamboor K Vishwanatha
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas.,Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas.,Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
41
|
Shologu N, Szegezdi E, Lowery A, Kerin M, Pandit A, Zeugolis DI. Recreating complex pathophysiologies in vitro with extracellular matrix surrogates for anticancer therapeutics screening. Drug Discov Today 2016; 21:1521-1531. [DOI: 10.1016/j.drudis.2016.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/17/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022]
|
42
|
Biochemical and biomechanical drivers of cancer cell metastasis, drug response and nanomedicine. Drug Discov Today 2016; 21:1489-1494. [PMID: 27238384 DOI: 10.1016/j.drudis.2016.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/11/2016] [Accepted: 05/18/2016] [Indexed: 12/27/2022]
Abstract
Metastasis, drug resistance and recurrence in cancer are regulated by the tumor microenvironment. This review describes recent advances in understanding how cancer cells respond to extracellular environmental cues via integrins, how to build engineered microenvironments to study these interactions in vitro and how nanomaterials can be used to detect and target tumor microenvironments.
Collapse
|
43
|
Holle AW, Young JL, Spatz JP. In vitro cancer cell-ECM interactions inform in vivo cancer treatment. Adv Drug Deliv Rev 2016; 97:270-9. [PMID: 26485156 DOI: 10.1016/j.addr.2015.10.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/05/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023]
Abstract
The general progression of cancer drug development involves in vitro testing followed by safety and efficacy evaluation in clinical trials. Due to the expense of bringing candidate drugs to trials, in vitro models of cancer cells and tumor biology are required to screen drugs. There are many examples of drugs exhibiting cytotoxic behavior in cancer cells in vitro but losing efficacy in vivo, and in many cases, this is the result of poorly understood chemoresistant effects conferred by the cancer microenvironment. To address this, improved methods for culturing cancer cells in biomimetic scaffolds have been developed; along the way, a great deal about the nature of cancer cell-extracellular matrix (ECM) interactions has been discovered. These discoveries will continue to be leveraged both in the development of novel drugs targeting these interactions and in the fabrication of biomimetic substrates for efficient cancer drug screening in vitro.
Collapse
|
44
|
Barney LE, Jansen LE, Polio SR, Galarza S, Lynch ME, Peyton SR. The Predictive Link between Matrix and Metastasis. Curr Opin Chem Eng 2016; 11:85-93. [PMID: 26942108 DOI: 10.1016/j.coche.2016.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer spread (metastasis) is responsible for 90% of cancer-related fatalities. Informing patient treatment to prevent metastasis, or kill all cancer cells in a patient's body before it becomes metastatic is extremely powerful. However, aggressive treatment for all non-metastatic patients is detrimental, both for quality of life concerns, and the risk of kidney or liver-related toxicity. Knowing when and where a patient has metastatic risk could revolutionize patient treatment and care. In this review, we attempt to summarize the key work of engineers and quantitative biologists in developing strategies and model systems to predict metastasis, with a particular focus on cell interactions with the extracellular matrix (ECM), as a tool to predict metastatic risk and tropism.
Collapse
Affiliation(s)
- L E Barney
- Department of Chemical Engineering, University of Massachusetts, Amherst Amherst, MA 01003
| | - L E Jansen
- Department of Chemical Engineering, University of Massachusetts, Amherst Amherst, MA 01003
| | - S R Polio
- Department of Chemical Engineering, University of Massachusetts, Amherst Amherst, MA 01003
| | - S Galarza
- Department of Chemical Engineering, University of Massachusetts, Amherst Amherst, MA 01003
| | - M E Lynch
- Department of Chemical Engineering, University of Massachusetts, Amherst Amherst, MA 01003; Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst Amherst, MA 01003
| | - S R Peyton
- Department of Chemical Engineering, University of Massachusetts, Amherst Amherst, MA 01003
| |
Collapse
|
45
|
Gu J, Chen J, Feng J, Liu Y, Xue Q, Mao G, Gai L, Lu X, Zhang R, Cheng J, Hu Y, Shao M, Shen H, Huang J. Overexpression of ADAMTS5 can regulate the migration and invasion of non-small cell lung cancer. Tumour Biol 2016; 37:8681-9. [PMID: 26738863 DOI: 10.1007/s13277-015-4573-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/01/2015] [Indexed: 01/01/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the major cause of cancer-related lethality among human cancer patients globally, and the poor prognosis of this cancer is mainly explained by metastasis, so it is essential to find out the molecule mechanisms and a novel therapeutic for NSCLC. A disintegrin and metalloprotease with thrombospondin motif 5 (ADAMTS5) belongs to the protease family. It has been reported to participate in tumor migration and invasion. In this study, we showed that the expression of ADAMTS5 was higher in lung cancer tissues by Western blot. The immunohistochemistry analysis was performed in 140 NSCLC cases, and the result indicated that ADAMTS5 was significantly associated with clinical pathologic variables. The Kaplan-Meier curve showed that the high expression of ADAMTS5 was related to poor prognosis of lung cancer patients. Wound healing assays and transwell migration assays revealed that the high expression of ADAMTS5 promoted the migration and invasion of NSCLC. In a word, our findings suggest that ADAMTS5 can regulate the migration and invasion of NSCLC and it may be a useful target of therapy in NSCLC.
Collapse
Affiliation(s)
- Jun Gu
- Department of Respiratory, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Respiratory, Affiliated Hospital of Nantong University, Nantong, China
| | - Jie Chen
- Department of Oncology, Jiangyin People's Hospital, Jiangyin, China
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jian Feng
- Department of Respiratory, Affiliated Hospital of Nantong University, Nantong, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qun Xue
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Guoxin Mao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Ling Gai
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoning Lu
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Rui Zhang
- Department of Respiratory, Affiliated Hospital of Nantong University, Nantong, China
| | - Jialin Cheng
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanxia Hu
- Department of Respiratory, Affiliated Hospital of Nantong University, Nantong, China
| | - Mengting Shao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hong Shen
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jianan Huang
- Department of Respiratory, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
46
|
|
47
|
Park KM, Gerecht S. Polymeric hydrogels as artificial extracellular microenvironments for cancer research. Eur Polym J 2015. [DOI: 10.1016/j.eurpolymj.2015.06.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
48
|
Moraes C. The Discovery Channel: microfluidics and microengineered systems in drug screening. Integr Biol (Camb) 2015; 7:285-8. [DOI: 10.1039/c5ib90004b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We highlight exciting findings and promising approaches in the recent literature in which researchers integrate advanced micro-engineering, design, and analytical strategies to improve the relevance and utility of high-throughput screening in the drug discovery pipeline.
Collapse
|