1
|
Rubí-Sans G, Nyga A, Mateos-Timoneda MA, Engel E. Substrate stiffness-dependent activation of Hippo pathway in cancer associated fibroblasts. BIOMATERIALS ADVANCES 2025; 166:214061. [PMID: 39406156 DOI: 10.1016/j.bioadv.2024.214061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/20/2024] [Accepted: 10/06/2024] [Indexed: 11/13/2024]
Abstract
The tumor microenvironment (TME) comprises a heterogenous cell population within a complex three-dimensional (3D) extracellular matrix (ECM). Stromal cells within this TME are altered by signaling cues from cancer cells to support uncontrolled tumor growth and invasion events. Moreover, the ECM also plays a fundamental role in tumor development through pathological remodeling, stiffening and interaction with TME cells. In healthy tissues, Hippo signaling pathway actively contributes to tissue growth, cell proliferation and apoptosis. However, in cancer, the Hippo signaling pathway is highly dysregulated, leading to nuclear translocation of the YAP/TAZ complex, which directly contributes to uncontrolled cell proliferation and tissue growth, and ECM remodeling and stiffening processes. Here, we compare the effect of increasing cell culture substrate stiffness, derived from tumor progression, upon the dysregulation of the Hippo signaling pathway in colorectal cancer-associated fibroblasts (CAFs) and normal colorectal fibroblasts (NFs). We correlate the dysregulation of Hippo pathway with the magnitude of the traction forces exerted by healthy and malignant stromal cells. We found that ECM stiffening is crucial in Hippo pathway dysregulation in CAFs, but not in normal fibroblasts.
Collapse
Affiliation(s)
- Gerard Rubí-Sans
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Agata Nyga
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.
| | - Miguel A Mateos-Timoneda
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain; IMEM-BRT group, Department of Materials Science, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain.
| |
Collapse
|
2
|
Behera SA, Nanda B, Achary PGR. Recent advancements and challenges in 3D bioprinting for cancer applications. BIOPRINTING 2024; 43:e00357. [DOI: 10.1016/j.bprint.2024.e00357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Sreedasyam R, Wilson BG, Ferrandez PR, Botvinick EL, Venugopalan V. An optical system for cellular mechanostimulation in 3D hydrogels. Acta Biomater 2024; 189:439-448. [PMID: 39368720 DOI: 10.1016/j.actbio.2024.09.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
We introduce a method utilizing single laser-generated cavitation bubbles to stimulate cellular mechanotransduction in dermal fibroblasts embedded within 3D hydrogels. We demonstrate that fibroblasts embedded in either amorphous or fibrillar hydrogels engage in Ca2+ signaling following exposure to an impulsive mechanical stimulus provided by a single 250 µm diameter laser-generated cavitation bubble. We find that the spatial extent of the cellular signaling is larger for cells embedded within a fibrous collagen hydrogel as compared to those embedded within an amorphous polyvinyl alcohol polymer (SLO-PVA) hydrogel. Additionally, for fibroblasts embedded in collagen, we find an increased range of cellular mechanosensitivity for cells that are polarized relative to the radial axis as compared to the circumferential axis. By contrast, fibroblasts embedded within SLO-PVA did not display orientation-dependent mechanosensitivity. Fibroblasts embedded in hydrogels and cultured in calcium-free media did not show cavitation-induced mechanotransduction; implicating calcium signaling based on transmembrane Ca2+ transport. This study demonstrates the utility of single laser-generated cavitation bubbles to provide local non-invasive impulsive mechanical stimuli within 3D hydrogel tissue models with concurrent imaging using optical microscopy. STATEMENT OF SIGNIFICANCE: Currently, there are limited methods for the non-invasive real-time assessment of cellular sensitivity to mechanical stimuli within 3D tissue scaffolds. We describe an original approach that utilizes a pulsed laser microbeam within a standard laser scanning microscope system to generate single cavitation bubbles to provide impulsive mechanostimulation to cells within 3D fibrillar and amorphous hydrogels. Using this technique, we measure the cellular mechanosensitivity of primary human dermal fibroblasts embedded in amorphous and fibrillar hydrogels, thereby providing a useful method to examine cellular mechanotransduction in 3D biomaterials. Moreover, the implementation of our method within a standard optical microscope makes it suitable for broad adoption by cellular mechanotransduction researchers and opens the possibility of high-throughput evaluation of biomaterials with respect to cellular mechanosignaling.
Collapse
Affiliation(s)
- Rahul Sreedasyam
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Bryce G Wilson
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States
| | - Patricia R Ferrandez
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| | - Vasan Venugopalan
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| |
Collapse
|
4
|
Yadav R, Sharma A, Dahiya D, Bal A, Bhatia A. Comparative morphology of tumour microenvironment in claudin-low and claudin-high breast cancers. Pathol Res Pract 2024; 261:155502. [PMID: 39079385 DOI: 10.1016/j.prp.2024.155502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/18/2024]
Abstract
BACKGROUND Claudin-low breast cancers (BCs) exhibit more aggressive behaviour compared to claudin-high types. Claudin-low BCs are often characterized by features such as a higher grade, enrichment of stemness characteristics, and a propensity for metastasis. Tumour microenvironment (TME) defined as the intricate network of surrounding cells, blood vessels, and extracellular matrix components influences the behaviour of cancer cells within the breast tissue. Understanding the TME is crucial for comprehending the aggressive characteristics of claudin-low BCs. METHODS In this study, we have studied the morphology of immune and non-immune TME using Haematoxylin and eosin (H&E)-stained slides of 15 claudin-low and 12 claudin-high tissue samples of BC. RESULTS TME of claudin-low BCs was observed to have a significantly higher frequency of retraction clefts (66.6 %; n = 10/15), immature desmoplastic response (40 %; n = 6/15), higher stromal cellularity (60 %; n = 9/15); and fibroblastic proliferation (53.3 %; n = 8/15) with a low prevalence of elastosis (66.6 %; n = 10/15). The immune microenvironment revealed a higher frequency of total (80 %; n = 12/15) as well as stromal (86.67 %; n = 13/15) and intra-tumoural TILs (60 %; n = 9/15) in them. CONCLUSION The above morphology-based study revealed that claudin-low tumours have unique immune and non-immune TME as compared to claudin-high tumours. Future studies exploring the molecular correlates of each of the above morphological features can help in identifying novel therapeutic targets for the treatment of claudin-low BCs.
Collapse
Affiliation(s)
- Reena Yadav
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Aditti Sharma
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Divya Dahiya
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Amanjit Bal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
5
|
Stoian A, Adil A, Biniazan F, Haykal S. Two Decades of Advances and Limitations in Organ Recellularization. Curr Issues Mol Biol 2024; 46:9179-9214. [PMID: 39194760 DOI: 10.3390/cimb46080543] [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: 07/31/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
The recellularization of tissues after decellularization is a relatively new technology in the field of tissue engineering (TE). Decellularization involves removing cells from a tissue or organ, leaving only the extracellular matrix (ECM). This can then be recellularized with new cells to create functional tissues or organs. The first significant mention of recellularization in decellularized tissues can be traced to research conducted in the early 2000s. One of the landmark studies in this field was published in 2008 by Ott, where researchers demonstrated the recellularization of a decellularized rat heart with cardiac cells, resulting in a functional organ capable of contraction. Since then, other important studies have been published. These studies paved the way for the widespread application of recellularization in TE, demonstrating the potential of decellularized ECM to serve as a scaffold for regenerating functional tissues. Thus, although the concept of recellularization was initially explored in previous decades, these studies from the 2000s marked a major turning point in the development and practical application of the technology for the recellularization of decellularized tissues. The article reviews the historical advances and limitations in organ recellularization in TE over the last two decades.
Collapse
Affiliation(s)
- Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Aisha Adil
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
- Reconstructive Oncology, Division of Plastic and Reconstructive Surgery, Smilow Cancer Hospital, Yale, New Haven, CT 06519, USA
| |
Collapse
|
6
|
Liu Y, Okesola BO, Osuna de la Peña D, Li W, Lin M, Trabulo S, Tatari M, Lawlor RT, Scarpa A, Wang W, Knight M, Loessner D, Heeschen C, Mata A, Pearce OMT. A Self-Assembled 3D Model Demonstrates How Stiffness Educates Tumor Cell Phenotypes and Therapy Resistance in Pancreatic Cancer. Adv Healthc Mater 2024; 13:e2301941. [PMID: 38471128 PMCID: PMC11468796 DOI: 10.1002/adhm.202301941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/16/2024] [Indexed: 03/14/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense and stiff extracellular matrix (ECM) associated with tumor progression and therapy resistance. To further the understanding of how stiffening of the tumor microenvironment (TME) contributes to aggressiveness, a three-dimensional (3D) self-assembling hydrogel disease model is developed based on peptide amphiphiles (PAs, PA-E3Y) designed to tailor stiffness. The model displays nanofibrous architectures reminiscent of native TME and enables the study of the invasive behavior of PDAC cells. Enhanced tuneability of stiffness is demonstrated by interacting thermally annealed aqueous solutions of PA-E3Y (PA-E3Yh) with divalent cations to create hydrogels with mechanical properties and ultrastructure similar to native tumor ECM. It is shown that stiffening of PA-E3Yh hydrogels to levels found in PDAC induces ECM deposition, promotes epithelial-to-mesenchymal transition (EMT), enriches CD133+/CXCR4+ cancer stem cells (CSCs), and subsequently enhances drug resistance. The findings reveal how a stiff 3D environment renders PDAC cells more aggressive and therefore more faithfully recapitulates in vivo tumors.
Collapse
Affiliation(s)
- Ying Liu
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Babatunde O. Okesola
- School of Life SciencesFaculty of Medicine and Health SciencesUniversity of NottinghamNottinghamNG7 2RDUK
| | - David Osuna de la Peña
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Weiqi Li
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Meng‐Lay Lin
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Sara Trabulo
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Marianthi Tatari
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Rita T. Lawlor
- Department of Diagnostics and Public HealthSection of PathologyUniversity of VeronaVerona37134Italy
- ARC‐NetApplied Research on Cancer CentreUniversity of VeronaVerona37134Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public HealthSection of PathologyUniversity of VeronaVerona37134Italy
- ARC‐NetApplied Research on Cancer CentreUniversity of VeronaVerona37134Italy
| | - Wen Wang
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Martin Knight
- Centre for BioengineeringSchool of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
- Centre for Predictive in vitro ModelsQueen Mary University of LondonLondonE1 4NSUK
| | - Daniela Loessner
- Barts Cancer InstituteQueen Mary University of LondonLondonEC1M 6BQUK
- Department of Chemical and Biological EngineeringFaculty of EngineeringMonash UniversityMelbourneVIC3800Australia
- Department of Materials Science and EngineeringFaculty of EngineeringMonash UniversityMelbourneVIC3800Australia
- Department of Anatomy and Developmental BiologyFaculty of MedicineNursing and Health SciencesMonash UniversityMelbourneVIC3800Australia
| | - Christopher Heeschen
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute – FPO – IRCCSCandiolo (TO)10060Italy
| | - Alvaro Mata
- School of PharmacyUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
- Biodiscovery InstituteUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
- Department of Chemical and Environmental EngineeringUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
| | | |
Collapse
|
7
|
Isogai T, Murali VS, Zhou F, Wang X, Rajendran D, Perez-Castro L, Venkateswaran N, Conacci-Sorrell M, Danuser G. Anchorage-independent cell proliferation promoted by fascin's F-actin bundling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592404. [PMID: 38746129 PMCID: PMC11092747 DOI: 10.1101/2024.05.04.592404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The actin filament (F-actin) bundling protein fascin-1 is highly enriched in many metastatic cancers. Fascin's contribution to metastasis have been ascribed to its enhancement of cell migration and invasion. However, mouse genetic studies clearly point to functions also in tumorigenesis, yet without mechanistic underpinnings. Here, we show that fascin expression promotes the formation of a non-canonical signaling complex that enables anchorage-independent proliferation. This complex shares similarities to focal adhesions and we refer to them as pseudo-adhesion signaling scaffolds (PASS). PASS are enriched with tyrosine phosphorylated proteins and require fascin's F-actin-bundling activity for its assembly. PASS serve as hubs for the Rac1/PAK/JNK proliferation signaling axis, driven by PASS-associated Rac-specific GEFs. Experimental disruption of either fascin or RacGEF function abrogates sustained proliferation of aggressive cancers in vitro and in vivo . These results add a new molecular element to the growing arsenal of metabolic and oncogenic signaling programs regulated by the cytoskeleton architecture.
Collapse
|
8
|
Khine YY, Nguyen H, Afolabi F, Lin CC. Fast-relaxing hydrogels with reversibly tunable mechanics for dynamic cancer cell culture. BIOMATERIALS ADVANCES 2024; 159:213829. [PMID: 38531258 PMCID: PMC11075809 DOI: 10.1016/j.bioadv.2024.213829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024]
Abstract
The mechanics of the tumor microenvironment (TME) significantly impact disease progression and the efficacy of anti-cancer therapeutics. While it is recognized that advanced in vitro cancer models will benefit cancer research, none of the current engineered extracellular matrices (ECM) adequately recapitulate the highly dynamic TME. Through integrating reversible boronate-ester bonding and dithiolane ring-opening polymerization, we fabricated synthetic polymer hydrogels with tumor-mimetic fast relaxation and reversibly tunable elastic moduli. Importantly, the crosslinking and dynamic stiffening of matrix mechanics were achieved in the absence of a photoinitiator, often the source of cytotoxicity. Central to this strategy was Poly(PEGA-co-LAA-co-AAPBA) (PELA), a highly defined polymer synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization. PELA contains dithiolane for initiator-free gel crosslinking, stiffening, and softening, as well as boronic acid for complexation with diol-containing polymers to give rise to tunable viscoelasticity. PELA hydrogels were highly cytocompatible for dynamic culture of patient-derived pancreatic cancer cells. It was found that the fast-relaxing matrix induced mesenchymal phenotype of cancer cells, and dynamic matrix stiffening restricted tumor spheroid growth. Moreover, this new dynamic viscoelastic hydrogel system permitted sequential stiffening and softening to mimic the physical changes of TME.
Collapse
Affiliation(s)
- Yee Yee Khine
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Han Nguyen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Favour Afolabi
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
| |
Collapse
|
9
|
Srbova L, Arasalo O, Lehtonen AJ, Pokki J. Measuring mechanical cues for modeling the stromal matrix in 3D cell cultures. SOFT MATTER 2024; 20:3483-3498. [PMID: 38587658 DOI: 10.1039/d3sm01425h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
A breast-cancer tumor develops within a stroma, a tissue where a complex extracellular matrix surrounds cells, mediating the cancer progression through biomechanical and -chemical cues. Current materials partially mimic the stromal matrix in 3D cell cultures but methods for measuring the mechanical properties of the matrix at cell-relevant-length scales and stromal-stiffness levels are lacking. Here, to address this gap, we developed a characterization approach that employs probe-based microrheometry and Bayesian modeling to quantify length-scale-dependent mechanics and mechanical heterogeneity as in the stromal matrix. We examined the interpenetrating network (IPN) composed of alginate scaffolds (for adjusting mechanics) and type-1 collagen (a stromal-matrix constituent). We analyzed viscoelasticity: absolute-shear moduli (stiffness/elasticity) and phase angles (viscous and elastic characteristics). We determined the relationship between microrheometry and rheometry information. Microrheometry reveals lower stiffness at cell-relevant scales, compared to macroscale rheometry, with dependency on the length scale (10 to 100 μm). These data show increasing IPN stiffness with crosslinking until saturation (≃15 mM of Ca2+). Furthermore, we report that IPN stiffness can be adjusted by modulating collagen concentration and interconnectivity (by polymerization temperature). The IPNs are heterogeneous structurally (in SEM) and mechanically. Interestingly, increased alginate crosslinking changes IPN heterogeneity in stiffness but not in phase angle, until the saturation. In contrast, such changes are undetectable in alginate scaffolds. Our nonlinear viscoelasticity analysis at tumor-cell-exerted strains shows that only the softer IPNs stiffen with strain, like the stromal-collagen constituent. In summary, our approach can quantify the stromal-matrix-related viscoelasticity and is likely applicable to other materials in 3D culture.
Collapse
Affiliation(s)
- Linda Srbova
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| | - Ossi Arasalo
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| | - Arttu J Lehtonen
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| | - Juho Pokki
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| |
Collapse
|
10
|
Hadzipasic M, Zhang S, Huang Z, Passaro R, Sten MS, Shankar GM, Nia HT. Emergence of nanoscale viscoelasticity from single cancer cells to established tumors. Biomaterials 2024; 305:122431. [PMID: 38169188 PMCID: PMC10837793 DOI: 10.1016/j.biomaterials.2023.122431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
Tumors are complex materials whose physical properties dictate growth and treatment outcomes. Recent evidence suggests time-dependent physical properties, such as viscoelasticity, are crucial, distinct mechanical regulators of cancer progression and malignancy, yet the genesis and consequences of tumor viscoelasticity are poorly understood. Here, using Wide-bandwidth AFM-based ViscoElastic Spectroscopy (WAVES) coupled with mathematical modeling, we probe the origins of tumor viscoelasticity. From single carcinoma cells to increasingly sized carcinoma spheroids to established tumors, we describe a stepwise evolution of dynamic mechanical properties that create a nanorheological signature of established tumors: increased stiffness, decreased rate-dependent stiffening, and reduced energy dissipation. We dissect this evolution of viscoelasticity by scale, and show established tumors use fluid-solid interactions as the dominant mechanism of mechanical energy dissipation as opposed to fluid-independent intrinsic viscoelasticity. Additionally, we demonstrate the energy dissipation mechanism in spheroids and established tumors is negatively correlated with the cellular density, and this relationship strongly depends on an intact actin cytoskeleton. These findings define an emergent and targetable signature of the physical tumor microenvironment, with potential for deeper understanding of tumor pathophysiology and treatment strategies.
Collapse
Affiliation(s)
- Muhamed Hadzipasic
- Department of Biomedical Engineering, Boston University Boston, MA, USA; Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School Boston, Massachusetts, USA
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Zhuoying Huang
- Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Rachel Passaro
- Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Margaret S Sten
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School Boston, Massachusetts, USA
| | - Ganesh M Shankar
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School Boston, Massachusetts, USA
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University Boston, MA, USA.
| |
Collapse
|
11
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
12
|
Sousa de Almeida M, Lee A, Itel F, Maniura-Weber K, Petri-Fink A, Rothen-Rutishauser B. The Effect of Substrate Properties on Cellular Behavior and Nanoparticle Uptake in Human Fibroblasts and Epithelial Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:342. [PMID: 38392715 PMCID: PMC10892529 DOI: 10.3390/nano14040342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024]
Abstract
The delivery of nanomedicines into cells holds enormous therapeutic potential; however little is known regarding how the extracellular matrix (ECM) can influence cell-nanoparticle (NP) interactions. Changes in ECM organization and composition occur in several pathophysiological states, including fibrosis and tumorigenesis, and may contribute to disease progression. We show that the physical characteristics of cellular substrates, that more closely resemble the ECM in vivo, can influence cell behavior and the subsequent uptake of NPs. Electrospinning was used to create two different substrates made of soft polyurethane (PU) with aligned and non-aligned nanofibers to recapitulate the ECM in two different states. To investigate the impact of cell-substrate interaction, A549 lung epithelial cells and MRC-5 lung fibroblasts were cultured on soft PU membranes with different alignments and compared against stiff tissue culture plastic (TCP)/glass. Both cell types could attach and grow on both PU membranes with no signs of cytotoxicity but with increased cytokine release compared with cells on the TCP. The uptake of silica NPs increased more than three-fold in fibroblasts but not in epithelial cells cultured on both membranes. This study demonstrates that cell-matrix interaction is substrate and cell-type dependent and highlights the importance of considering the ECM and tissue mechanical properties when designing NPs for effective cell targeting and treatment.
Collapse
Affiliation(s)
- Mauro Sousa de Almeida
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
| | - Aaron Lee
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
- Department of Bioengineering, Imperial College London, South Kensington, London SW7 2BP, UK
| | - Fabian Itel
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biomimetic Membranes and Textiles, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland;
| | - Katharina Maniura-Weber
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biointerfaces, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland;
| | - Alke Petri-Fink
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Barbara Rothen-Rutishauser
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
| |
Collapse
|
13
|
Han S, Zhang M, Qu X, Wu Z, Huang Z, Hu Y, Li Y, Cui L, Si L, Liu J, Shao Y. SOX10 deficiency-mediated LAMB3 upregulation determines the invasiveness of MAPKi-resistant melanoma. Oncogene 2024; 43:434-446. [PMID: 38102338 DOI: 10.1038/s41388-023-02917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
Melanoma that develops adaptive resistance to MAPK inhibitors (MAPKi) through transcriptional reprograming-mediated phenotype switching is associated with enhanced metastatic potential, yet the underlying mechanism of this improved invasiveness has not been fully elucidated. In this study, we show that MAPKi-resistant melanoma cells are more motile and invasive than the parental cells. We further show that LAMB3, a β subunit of the extracellular matrix protein laminin-332 is upregulated in MAPKi-resistant melanoma cells and that the LAMB3-Integrin α3/α6 signaling mediates the motile and invasive phenotype of resistant cells. In addition, we demonstrate that SOX10 deficiency in MAPKi-resistant melanoma cells drives LAMB3 upregulation through TGF-β signaling. Transcriptome profiling and functional studies further reveal a FAK/MMPs axis mediates the pro-invasiveness effect of LAMB3. Using a mouse lung metastasis model, we demonstrate LAMB3 depletion inhibits the metastatic potential of MAPKi-resistant cells in vivo. In summary, this study identifies a SOX10low/TGF-β/LAMB3/FAK/MMPs signaling pathway that determines the migration and invasion properties of MAPKi-resistant melanoma cells and provide rationales for co-targeting LAMB3 to curb the metastasis of melanoma cells in targeted therapy.
Collapse
Affiliation(s)
- Shujun Han
- Frontier Institute of Science and Technology, and Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Mo Zhang
- Frontier Institute of Science and Technology, and Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaoyan Qu
- Frontier Institute of Science and Technology, and Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zihao Wu
- Frontier Institute of Science and Technology, and Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zongguan Huang
- Frontier Institute of Science and Technology, and Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yiming Hu
- Department of Dermatology, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Li
- Department of Dermatology, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Lanlan Cui
- Department of Dermatology, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing, 100142, China
| | - Jiankang Liu
- Frontier Institute of Science and Technology, and Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China.
| | - Yongping Shao
- Frontier Institute of Science and Technology, and Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
- Department of Dermatology, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
14
|
Samaržija I. The Potential of Extracellular Matrix- and Integrin Adhesion Complex-Related Molecules for Prostate Cancer Biomarker Discovery. Biomedicines 2023; 12:79. [PMID: 38255186 PMCID: PMC10813710 DOI: 10.3390/biomedicines12010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is among the top five cancer types according to incidence and mortality. One of the main obstacles in prostate cancer management is the inability to foresee its course, which ranges from slow growth throughout years that requires minimum or no intervention to highly aggressive disease that spreads quickly and resists treatment. Therefore, it is not surprising that numerous studies have attempted to find biomarkers of prostate cancer occurrence, risk stratification, therapy response, and patient outcome. However, only a few prostate cancer biomarkers are used in clinics, which shows how difficult it is to find a novel biomarker. Cell adhesion to the extracellular matrix (ECM) through integrins is among the essential processes that govern its fate. Upon activation and ligation, integrins form multi-protein intracellular structures called integrin adhesion complexes (IACs). In this review article, the focus is put on the biomarker potential of the ECM- and IAC-related molecules stemming from both body fluids and prostate cancer tissue. The processes that they are involved in, such as tumor stiffening, bone turnover, and communication via exosomes, and their biomarker potential are also reviewed.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
15
|
G K AV, Gogoi G, Kachappilly MC, Rangarajan A, Pandya HJ. Label-free multimodal electro-thermo-mechanical (ETM) phenotyping as a novel biomarker to differentiate between normal, benign, and cancerous breast biopsy tissues. J Biol Eng 2023; 17:68. [PMID: 37957665 PMCID: PMC10644568 DOI: 10.1186/s13036-023-00388-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Technologies for quick and label-free diagnosis of malignancies from breast tissues have the potential to be a significant adjunct to routine diagnostics. The biophysical phenotypes of breast tissues, such as its electrical, thermal, and mechanical properties (ETM), have the potential to serve as novel markers to differentiate between normal, benign, and malignant tissue. RESULTS We report a system-of-biochips (SoB) integrated into a semi-automated mechatronic system that can characterize breast biopsy tissues using electro-thermo-mechanical sensing. The SoB, fabricated on silicon using microfabrication techniques, can measure the electrical impedance (Z), thermal conductivity (K), mechanical stiffness (k), and viscoelastic stress relaxation (%R) of the samples. The key sensing elements of the biochips include interdigitated electrodes, resistance temperature detectors, microheaters, and a micromachined diaphragm with piezoresistive bridges. Multi-modal ETM measurements performed on formalin-fixed tumour and adjacent normal breast biopsy samples from N = 14 subjects were able to differentiate between invasive ductal carcinoma (malignant), fibroadenoma (benign), and adjacent normal (healthy) tissues with a root mean square error of 0.2419 using a Gaussian process classifier. Carcinoma tissues were observed to have the highest mean impedance (110018.8 ± 20293.8 Ω) and stiffness (0.076 ± 0.009 kNm-1) and the lowest thermal conductivity (0.189 ± 0.019 Wm-1 K-1) amongst the three groups, while the fibroadenoma samples had the highest percentage relaxation in normalized load (47.8 ± 5.12%). CONCLUSIONS The work presents a novel strategy to characterize the multi-modal biophysical phenotype of breast biopsy tissues to aid in cancer diagnosis from small-sized tumour samples. The methodology envisions to supplement the existing technology gap in the analysis of breast tissue samples in the pathology laboratories to aid the diagnostic workflow.
Collapse
Affiliation(s)
- Anil Vishnu G K
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Gayatri Gogoi
- Department of Pathology, Assam Medical College, Dibrugarh, Assam, 786002, India
| | - Midhun C Kachappilly
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Annapoorni Rangarajan
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Hardik J Pandya
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, Karnataka, 560012, India.
- Centre for Product Design and Manufacturing, Indian Institute of Science, Bangalore, Karnataka, 560012, India.
| |
Collapse
|
16
|
Gao Z, Guo J, Gou B, Gu Z, Jia T, Ma S, Jiang L, Liu W, Zhou L, Gu Q. Microcarriers promote the through interface movement of mouse trophoblast stem cells by regulating stiffness. Bioact Mater 2023; 28:196-205. [PMID: 37250864 PMCID: PMC10220236 DOI: 10.1016/j.bioactmat.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/20/2023] [Accepted: 05/09/2023] [Indexed: 05/31/2023] Open
Abstract
Mechanical force is crucial in the whole process of embryonic development. However, the role of trophoblast mechanics during embryo implantation has rarely been studied. In this study, we constructed a model to explore the effect of stiffness changes in mouse trophoblast stem cells (mTSCs) on implantation: microcarrier was prepared by sodium alginate using a droplet microfluidics system, and mTSCs were attached to the microcarrier surface with laminin modifications, called T(micro). Compared with the spheroid, formed by the self-assembly of mTSCs (T(sph)), we could regulate the stiffness of the microcarrier, making the Young's modulus of mTSCs (367.70 ± 79.81 Pa) similar to that of the blastocyst trophoblast ectoderm (432.49 ± 151.90 Pa). Moreover, T(micro) contributes to improve the adhesion rate, expansion area and invasion depth of mTSCs. Further, T(micro) was highly expressed in tissue migration-related genes due to the activation of the Rho-associated coiled-coil containing protein kinase (ROCK) pathway at relatively similar modulus of trophoblast. Overall, our study explores the embryo implantation process with a new perspective, and provides theoretical support for understanding the effect of mechanics on embryo implantation.
Collapse
Affiliation(s)
- Zili Gao
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jia Guo
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Bo Gou
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhen Gu
- Department of Chemistry and Biological Engineering, University of Science and Technology, Beijing, 100083, PR China
| | - Tan Jia
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Sinan Ma
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- School of Life Sciences, Northeast Agricultural University, Harbin, 150030, PR China
| | - Liyuan Jiang
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- School of Life Sciences, Northeast Agricultural University, Harbin, 150030, PR China
| | - Wenli Liu
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
| | - Lixun Zhou
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
17
|
Li G, Cai J, Xie J, Dai Y. Extracellular fibrin promotes non-small cell lung cancer progression through integrin β1/PTEN/AKT signaling. Open Life Sci 2023; 18:20220716. [PMID: 37744455 PMCID: PMC10512450 DOI: 10.1515/biol-2022-0716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 09/26/2023] Open
Abstract
The extracellular matrix (ECM) has been strongly correlated with cancer progression in various tumor types. However, the specific mechanisms underlying ECM-associated tumor behaviors remain unclear. In this study, we found an enriched distribution of fibrin in tumor tissues obtained from high-grade non-small cell lung cancer (NSCLC) patients. For further investigation, we established an in vitro 3D culture system using fibrin gel and found that NSCLC cells grown in this system exhibited increased stemness and tumorigenesis. Mechanistically, we demonstrated that fibrin facilitated the activation of the phosphatase and tensin homolog (PTEN)/protein kinase B (AKT) signaling pathway through integrin β1. Furthermore, we found that blocking integrin β1 signals enhanced the tumor suppressive effects of chemotherapy, providing a novel approach for clinical therapy for NSCLC.
Collapse
Affiliation(s)
- Guilong Li
- Department of Cardiothoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian 363000, The People’s Republic of China
| | - Jiaying Cai
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian 363000, The People’s Republic of China
| | - Jianjun Xie
- Department of Radiotherapy, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian 363000, The People’s Republic of China
| | - Yizhi Dai
- Department of Cardiothoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian 363000, The People’s Republic of China
| |
Collapse
|
18
|
Luo Z, Wan R, Liu S, Feng X, Peng Z, Wang Q, Chen S, Shang X. Mechanisms of exercise in the treatment of lung cancer - a mini-review. Front Immunol 2023; 14:1244764. [PMID: 37691942 PMCID: PMC10483406 DOI: 10.3389/fimmu.2023.1244764] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Lung cancer constitutes a formidable menace to global health and well-being, as its incidence and mortality rate escalate at an alarming pace. In recent years, research has indicated that exercise has potential roles in both the prevention and treatment of lung cancer. However, the exact mechanism of the coordinating effect of exercise on lung cancer treatment is unclear, limiting the use of exercise in clinical practice. The purpose of this review is to explore the mechanisms through which exercise exerts its anticancer effects against lung cancer. This review will analyze the biological basis of exercise's anticancer effects on lung cancer, with a focus on aspects such as the tumor microenvironment, matrix regulation, apoptosis and angiogenesis. Finally, we will discuss future research directions and potential clinical applications.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shan Liu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Peng
- Department of Sports Medicine, Shanghai General Hospital, Shanghai, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiliang Shang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Guo KS, Brodsky AS. Tumor collagens predict genetic features and patient outcomes. NPJ Genom Med 2023; 8:15. [PMID: 37414817 DOI: 10.1038/s41525-023-00358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/14/2023] [Indexed: 07/08/2023] Open
Abstract
The extracellular matrix (ECM) is a critical determinant of tumor fate that reflects the output from myriad cell types in the tumor. Collagens constitute the principal components of the tumor ECM. The changing collagen composition in tumors along with their impact on patient outcomes and possible biomarkers remains largely unknown. The RNA expression of the 43 collagen genes from solid tumors in The Cancer Genome Atlas (TCGA) was clustered to classify tumors. PanCancer analysis revealed how collagens by themselves can identify the tissue of origin. Clustering by collagens in each cancer type demonstrated strong associations with survival, specific immunoenvironments, somatic gene mutations, copy number variations, and aneuploidy. We developed a machine learning classifier that predicts aneuploidy, and chromosome arm copy number alteration (CNA) status based on collagen expression alone with high accuracy in many cancer types with somatic mutations, suggesting a strong relationship between the collagen ECM context and specific molecular alterations. These findings have broad implications in defining the relationship between cancer-related genetic defects and the tumor microenvironment to improve prognosis and therapeutic targeting for patient care, opening new avenues of investigation to define tumor ecosystems.
Collapse
Affiliation(s)
- Kevin S Guo
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Alexander S Brodsky
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA.
| |
Collapse
|
20
|
Ahmed T. Biomaterial-based in vitro 3D modeling of glioblastoma multiforme. CANCER PATHOGENESIS AND THERAPY 2023; 1:177-194. [PMID: 38327839 PMCID: PMC10846340 DOI: 10.1016/j.cpt.2023.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/24/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2024]
Abstract
Adult-onset brain cancers, such as glioblastomas, are particularly lethal. People with glioblastoma multiforme (GBM) do not anticipate living for more than 15 months if there is no cure. The results of conventional treatments over the past 20 years have been underwhelming. Tumor aggressiveness, location, and lack of systemic therapies that can penetrate the blood-brain barrier are all contributing factors. For GBM treatments that appear promising in preclinical studies, there is a considerable rate of failure in phase I and II clinical trials. Unfortunately, access becomes impossible due to the intricate architecture of tumors. In vitro, bioengineered cancer models are currently being used by researchers to study disease development, test novel therapies, and advance specialized medications. Many different techniques for creating in vitro systems have arisen over the past few decades due to developments in cellular and tissue engineering. Later-stage research may yield better results if in vitro models that resemble brain tissue and the blood-brain barrier are used. With the use of 3D preclinical models made available by biomaterials, researchers have discovered that it is possible to overcome these limitations. Innovative in vitro models for the treatment of GBM are possible using biomaterials and novel drug carriers. This review discusses the benefits and drawbacks of 3D in vitro glioblastoma modeling systems.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| |
Collapse
|
21
|
Cameron AP, Gao S, Liu Y, Zhao CX. Impact of hydrogel biophysical properties on tumor spheroid growth and drug response. BIOMATERIALS ADVANCES 2023; 149:213421. [PMID: 37060634 DOI: 10.1016/j.bioadv.2023.213421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
The extracellular matrix (ECM) plays a critical role in regulating cell-matrix interactions during tumor progression. These interactions are due in large part to the biophysical properties responding to cancer cell interactions. Within in vitro models, the ECM is mimicked by hydrogels, which possess adjustable biophysical properties that are integral to tumor development. This work presents a systematic and comparative study on the impact of the biophysical properties of two widely used natural hydrogels, Matrigel and collagen gel, on tumor growth and drug response. The biophysical properties of Matrigel and collagen including complex modulus, loss tangent, diffusive permeability, and pore size, were characterised. Then the spheroid growth rates in these two hydrogels were monitored for spheroids with two different sizes (140 μm and 500 μm in diameters). An increased migratory growth was observed in the lower concentration of both the gels. The effect of spheroid incorporation within the hydrogel had a minimal impact on the hydrogel's complex modulus. Finally, 3D tumor models using different concentrations of hydrogels were applied for drug treatment using paclitaxel. Spheroids cultured in hydrogels with different concentrations showed different drug response, demonstrating the significant effect of the choice of hydrogels and their concentrations on the drug response results despite using the same spheroids. This study provides useful insights into the effect of hydrogel biophysical properties on spheroid growth and drug response and highlights the importance of hydrogel selection and in vitro model design.
Collapse
Affiliation(s)
- Anna P Cameron
- Australian institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Song Gao
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, Australia
| | - Yun Liu
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, Australia
| | - Chun-Xia Zhao
- Australian institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia; School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
22
|
Shou Y, Teo XY, Li X, Zhicheng L, Liu L, Sun X, Jonhson W, Ding J, Lim CT, Tay A. Dynamic Magneto-Softening of 3D Hydrogel Reverses Malignant Transformation of Cancer Cells and Enhances Drug Efficacy. ACS NANO 2023; 17:2851-2867. [PMID: 36633150 DOI: 10.1021/acsnano.2c11278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
High extracellular matrix stiffness is a prominent feature of malignant tumors associated with poor clinical prognosis. To elucidate mechanistic connections between increased matrix stiffness and tumor progression, a variety of hydrogel scaffolds with dynamic changes in stiffness have been developed. These approaches, however, are not biocompatible at high temperature, strong irradiation, and acidic/basic pH, often lack reversibility (can only stiffen and not soften), and do not allow study on the same cell population longitudinally. In this work, we develop a dynamic 3D magnetic hydrogel whose matrix stiffness can be wirelessly and reversibly stiffened and softened multiple times with different rates of change using an external magnet. With this platform, we found that matrix stiffness increased tumor malignancy including denser cell organization, epithelial-to-mesenchymal transition and hypoxia. More interestingly, these malignant transformations could be halted or reversed with matrix softening (i.e., mechanical rescue), to potentiate drug efficacy attributing to reduced solid stress from matrix and downregulation of cell mechano-transductors including YAP1. We propose that our platform can be used to deepen understanding of the impact of matrix softening on cancer biology, an important but rarely studied phenomenon.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Xin Yong Teo
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Le Zhicheng
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Ling Liu
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| | - Xinhong Sun
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Win Jonhson
- Department of Materials Science and Engineering, National University of Singapore, 117575, Singapore
| | - Jun Ding
- Department of Materials Science and Engineering, National University of Singapore, 117575, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
23
|
Sarkar M, Nguyen T, Gundre E, Ogunlusi O, El-Sobky M, Giri B, Sarkar TR. Cancer-associated fibroblasts: The chief architect in the tumor microenvironment. Front Cell Dev Biol 2023; 11:1089068. [PMID: 36793444 PMCID: PMC9923123 DOI: 10.3389/fcell.2023.1089068] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Stromal heterogeneity of tumor microenvironment (TME) plays a crucial role in malignancy and therapeutic resistance. Cancer-associated fibroblasts (CAFs) are one of the major players in tumor stroma. The heterogeneous sources of origin and subsequent impacts of crosstalk with breast cancer cells flaunt serious challenges before current therapies to cure triple-negative breast cancer (TNBC) and other cancers. The positive and reciprocal feedback of CAFs to induce cancer cells dictates their mutual synergy in establishing malignancy. Their substantial role in creating a tumor-promoting niche has reduced the efficacy of several anti-cancer treatments, including radiation, chemotherapy, immunotherapy, and endocrine therapy. Over the years, there has been an emphasis on understanding CAF-induced therapeutic resistance in order to enhance cancer therapy results. CAFs, in the majority of cases, employ crosstalk, stromal management, and other strategies to generate resilience in surrounding tumor cells. This emphasizes the significance of developing novel strategies that target particular tumor-promoting CAF subpopulations, which will improve treatment sensitivity and impede tumor growth. In this review, we discuss the current understanding of the origin and heterogeneity of CAFs, their role in tumor progression, and altering the tumor response to therapeutic agents in breast cancer. In addition, we also discuss the potential and possible approaches for CAF-mediated therapies.
Collapse
Affiliation(s)
- Mrinmoy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Tristan Nguyen
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Esheksha Gundre
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Olajumoke Ogunlusi
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Mohanad El-Sobky
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, English Bazar, India
| | - Tapasree Roy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
24
|
Owen JS, Clayton A, Pearson HB. Cancer-Associated Fibroblast Heterogeneity, Activation and Function: Implications for Prostate Cancer. Biomolecules 2022; 13:67. [PMID: 36671452 PMCID: PMC9856041 DOI: 10.3390/biom13010067] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The continuous remodeling of the tumor microenvironment (TME) during prostate tumorigenesis is emerging as a critical event that facilitates cancer growth, progression and drug-resistance. Recent advances have identified extensive communication networks that enable tumor-stroma cross-talk, and emphasized the functional importance of diverse, heterogeneous stromal fibroblast populations during malignant growth. Cancer-associated fibroblasts (CAFs) are a vital component of the TME, which mediate key oncogenic events including angiogenesis, immunosuppression, metastatic progression and therapeutic resistance, thus presenting an attractive therapeutic target. Nevertheless, how fibroblast heterogeneity, recruitment, cell-of-origin and differential functions contribute to prostate cancer remains to be fully delineated. Developing our molecular understanding of these processes is fundamental to developing new therapies and biomarkers that can ultimately improve clinical outcomes. In this review, we explore the current challenges surrounding fibroblast identification, discuss new mechanistic insights into fibroblast functions during normal prostate tissue homeostasis and tumorigenesis, and illustrate the diverse nature of fibroblast recruitment and CAF generation. We also highlight the promise of CAF-targeted therapies for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Jasmine S. Owen
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Aled Clayton
- Tissue Microenvironment Group, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
25
|
Lee M, Du H, Winer DA, Clemente-Casares X, Tsai S. Mechanosensing in macrophages and dendritic cells in steady-state and disease. Front Cell Dev Biol 2022; 10:1044729. [PMID: 36467420 PMCID: PMC9712790 DOI: 10.3389/fcell.2022.1044729] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Macrophages and dendritic cells are myeloid cells that play critical roles in immune responses. Macrophages help to maintain homeostasis through tissue regeneration and the clearance of dead cells, but also mediate inflammatory processes against invading pathogens. As the most potent antigen-presenting cells, dendritic cells are important in connecting innate to adaptive immune responses via activation of T cells, and inducing tolerance under physiological conditions. While it is known that macrophages and dendritic cells respond to biochemical cues in the microenvironment, the role of extracellular mechanical stimuli is becoming increasingly apparent. Immune cell mechanotransduction is an emerging field, where accumulating evidence suggests a role for extracellular physical cues coming from tissue stiffness in promoting immune cell recruitment, activation, metabolism and inflammatory function. Additionally, many diseases such as pulmonary fibrosis, cardiovascular disease, cancer, and cirrhosis are associated with changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, topography, and mechanical forces on macrophage and dendritic cell behavior under steady-state and pathophysiological conditions. In addition, we will also provide insight on molecular mediators and signaling pathways important in macrophage and dendritic cell mechanotransduction.
Collapse
Affiliation(s)
- Megan Lee
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Huixun Du
- Buck Institute for Research on Aging, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Daniel A. Winer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Xavier Clemente-Casares
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Sue Tsai
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
26
|
Dornhof J, Zieger V, Kieninger J, Frejek D, Zengerle R, Urban GA, Kartmann S, Weltin A. Bioprinting-based automated deposition of single cancer cell spheroids into oxygen sensor microelectrode wells. LAB ON A CHIP 2022; 22:4369-4381. [PMID: 36254669 DOI: 10.1039/d2lc00705c] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Three-dimensional (3D) cell agglomerates, such as microtissues, organoids, and spheroids, become increasingly relevant in biomedicine. They can provide in vitro models that recapitulate functions of the original tissue in the body and have applications in cancer research. For example, they are widely used in organ-on-chip systems. Microsensors can provide essential real-time information on cell metabolism as well as the reliability and quality of culture conditions. The combination of sensors and 3D cell cultures, especially single spheroids, is challenging in terms of reproducible formation, manipulation, and access to spheroids, precise positioning near sensors, and high cell-to-volume ratios to obtain meaningful biosignals in the most parallel approach possible. To overcome this challenge, we combined state-of-the-art bioprinting techniques to automatically print tumour spheroids directly into microwells of a chip-based electrochemical oxygen sensor array. We demonstrated highly accurate and reproducible spheroid formation (diameter of approx. 200 μm) and a spheroid deposition precision of 25 μm within a volume of 22 nl per droplet. Microstructures and hydrogel-coated microwells allowed the placement of single MCF-7 breast cancer spheroids close to the sensor electrodes. The microelectrode wells were sealed for oxygen measurements within a 55 nl volume for fast concentration changes. Accurate and stable amperometric oxygen sensor performance was demonstrated from atmospheric to anoxic regions. Cellular respiration rates from single tumour spheroids in the range of 450-850 fmol min-1 were determined, and alterations of cell metabolism upon drug exposure were shown. Our results uniquely combine bioprinting with 3D cell culture monitoring and demonstrate the much-needed effort for facilitation, parallelization, sensor integration, and drug delivery in 3D cell culture and organ-on-chip experiments. The workflow has a high degree of automation and potential for scalability. In order to achieve greater flexibility in the automation of spheroid formation and trapping, we employed a method based on drop-on-demand liquid handling systems, instead of the typical on-chip approach commonly used in microfluidics. Its relevance ranges from fundamental metabolic research over standardization of cell culture experiments and toxicological studies, to personalized medicine, e.g. patient-specific chemotherapy.
Collapse
Affiliation(s)
- Johannes Dornhof
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | - Viktoria Zieger
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Jochen Kieninger
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | | | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
- Hahn-Schickard, Freiburg, Germany
| | - Gerald A Urban
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | - Sabrina Kartmann
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
- Hahn-Schickard, Freiburg, Germany
| | - Andreas Weltin
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
27
|
Lee A, Sousa de Almeida M, Milinkovic D, Septiadi D, Taladriz-Blanco P, Loussert-Fonta C, Balog S, Bazzoni A, Rothen-Rutishauser B, Petri-Fink A. Substrate stiffness reduces particle uptake by epithelial cells and macrophages in a size-dependent manner through mechanoregulation. NANOSCALE 2022; 14:15141-15155. [PMID: 36205559 PMCID: PMC9585528 DOI: 10.1039/d2nr03792k] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/30/2022] [Indexed: 05/23/2023]
Abstract
Cells continuously exert forces on their environment and respond to changes in mechanical forces by altering their behaviour. Many pathologies such as cancer and fibrosis are hallmarked by dysregulation in the extracellular matrix, driving aberrant behaviour through mechanotransduction pathways. We demonstrate that substrate stiffness can be used to regulate cellular endocytosis of particles in a size-dependent fashion. Culture of A549 epithelial cells and J774A.1 macrophages on polystyrene/glass (stiff) and polydimethylsiloxane (soft) substrates indicated that particle uptake is increased up to six times for A549 and two times for macrophages when cells are grown in softer environments. Furthermore, we altered surface characteristics through the attachment of submicron-sized particles as a method to locally engineer substrate stiffness and topography to investigate the biomechanical changes which occurred within adherent epithelial cells, i.e. characterization of A549 cell spreading and focal adhesion maturation. Consequently, decreasing substrate rigidity and particle-based topography led to a reduction of focal adhesion size. Moreover, expression levels of Yes-associated protein were found to correlate with the degree of particle endocytosis. A thorough appreciation of the mechanical cues may lead to improved solutions to optimize nanomedicine approaches for treatment of cancer and other diseases with abnormal mechanosignalling.
Collapse
Affiliation(s)
- Aaron Lee
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Mauro Sousa de Almeida
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Daela Milinkovic
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Dedy Septiadi
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Patricia Taladriz-Blanco
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
- International Iberian Nanotechnology Laboratory (INL), Water Quality Group, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Céline Loussert-Fonta
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Sandor Balog
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Amelie Bazzoni
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| |
Collapse
|
28
|
Esfahani P, Sun B. Patterning ECM microstructure to investigate 3D cellular dynamics under multiplexed mechanochemical guidance. F1000Res 2022; 11:1071. [PMID: 37901154 PMCID: PMC10603315 DOI: 10.12688/f1000research.125171.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 10/31/2023] Open
Abstract
Background: Biochemical and biophysical factors jointly regulate the cellular dynamics in many physiological processes. It is therefore imperative to include multiplexed microenvironment cues when employing {in vitro} cell-based assays to model physiological processes. Methods: To meet this need, we have developed a modular platform of 3D cell culture, Modular Control of Microenvironment for Cell Migration and Culture Assay (MC33A), that incorporates directed chemical and mechanical cues in the forms of chemotaxis and contact guidance, respectively. Taking advantage of the functionalities of MC33A, we study the migration and morphology of breast cancer cells in 3D engineered extracellular matrix (ECM) following a serum gradient for chemotaxis. Results: We show that when chemotaxis is facilitated by contact guidance in the same direction as the serum gradient, cells demonstrate dimensional-reduction in their motility and highly elongated ellipsoidal shape. When the direction of ECM alignment diverges from the direction of serum gradient, chemotactic motion is significantly suppressed, and cells are generally more protrusive and rounded in their morphology. Conclusions: These examples demonstrate MC33A as a powerful tool for engineering complex microenvironments of cells that will advance the state-of-the-art of cell-based analysis in drug development, regenerative medicine, and other research areas in bioengineering.
Collapse
Affiliation(s)
| | - Bo Sun
- Physics, Oregon State University, Corvallis, OR, 97330, USA
| |
Collapse
|
29
|
Jurj A, Ionescu C, Berindan-Neagoe I, Braicu C. The extracellular matrix alteration, implication in modulation of drug resistance mechanism: friends or foes? J Exp Clin Cancer Res 2022; 41:276. [PMID: 36114508 PMCID: PMC9479349 DOI: 10.1186/s13046-022-02484-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022] Open
Abstract
The extracellular matrix (ECM) is an important component of the tumor microenvironment (TME), having several important roles related to the hallmarks of cancer. In cancer, multiple components of the ECM have been shown to be altered. Although most of these alterations are represented by the increased or decreased quantity of the ECM components, changes regarding the functional alteration of a particular ECM component or of the ECM as a whole have been described. These alterations can be induced by the cancer cells directly or by the TME cells, with cancer-associated fibroblasts being of particular interest in this regard. Because the ECM has this wide array of functions in the tumor, preclinical and clinical studies have assessed the possibility of targeting the ECM, with some of them showing encouraging results. In the present review, we will highlight the most relevant ECM components presenting a comprehensive description of their physical, cellular and molecular properties which can alter the therapy response of the tumor cells. Lastly, some evidences regarding important biological processes were discussed, offering a more detailed understanding of how to modulate altered signalling pathways and to counteract drug resistance mechanisms in tumor cells.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | - Calin Ionescu
- 7Th Surgical Department, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012, Cluj-Napoca, Romania
- Surgical Department, Municipal Hospital, 400139, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
- Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, 540139, Targu Mures, Romania.
| |
Collapse
|
30
|
Popovic A, Tartare-Deckert S. Role of extracellular matrix architecture and signaling in melanoma therapeutic resistance. Front Oncol 2022; 12:924553. [PMID: 36119516 PMCID: PMC9479148 DOI: 10.3389/fonc.2022.924553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/15/2022] [Indexed: 12/03/2022] Open
Abstract
The extracellular matrix (ECM) is critical for maintaining tissue homeostasis therefore its production, assembly and mechanical stiffness are highly regulated in normal tissues. However, in solid tumors, increased stiffness resulting from abnormal ECM structural changes is associated with disease progression, an increased risk of metastasis and poor survival. As a dynamic and key component of the tumor microenvironment, the ECM is becoming increasingly recognized as an important feature of tumors, as it has been shown to promote several hallmarks of cancer via biochemical and biomechanical signaling. In this regard, melanoma cells are highly sensitive to ECM composition, stiffness and fiber alignment because they interact directly with the ECM in the tumor microenvironment via cell surface receptors, secreted factors or enzymes. Importantly, seeing as the ECM is predominantly deposited and remodeled by myofibroblastic stromal fibroblasts, it is a key avenue facilitating their paracrine interactions with melanoma cells. This review gives an overview of melanoma and further describes the critical roles that ECM properties such as ECM remodeling, ECM-related proteins and stiffness play in cutaneous melanoma progression, tumor cell plasticity and therapeutic resistance. Finally, given the emerging importance of ECM dynamics in melanoma, future perspectives on therapeutic strategies to normalize the ECM in tumors are discussed.
Collapse
Affiliation(s)
- Ana Popovic
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Team Microenvironnement, Signaling and Cancer, Equipe Labellisée Ligue Contre le Cancer, Nice, France
| | - Sophie Tartare-Deckert
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Team Microenvironnement, Signaling and Cancer, Equipe Labellisée Ligue Contre le Cancer, Nice, France
| |
Collapse
|
31
|
Gupta VK, Chaudhuri O. Mechanical regulation of cell-cycle progression and division. Trends Cell Biol 2022; 32:773-785. [PMID: 35491306 PMCID: PMC9378598 DOI: 10.1016/j.tcb.2022.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
Abstract
Cell-cycle progression and division are fundamental biological processes in animal cells, and their biochemical regulation has been extensively studied. An emerging body of work has revealed how mechanical interactions of cells with their microenvironment in tissues, including with the extracellular matrix (ECM) and neighboring cells, also plays a crucial role in regulating cell-cycle progression and division. We review recent work on how cells interpret physical cues and alter their mechanics to promote cell-cycle progression and initiate cell division, and then on how dividing cells generate forces on their surrounding microenvironment to successfully divide. Finally, the article ends by discussing how force generation during division potentially contributes to larger tissue-scale processes involved in development and homeostasis.
Collapse
Affiliation(s)
- Vivek K Gupta
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA..
| |
Collapse
|
32
|
Habbit NL, Anbiah B, Anderson L, Suresh J, Hassani I, Eggert M, Brannen A, Davis J, Tian Y, Prabhakarpandian B, Panizzi P, Arnold RD, Lipke EA. Tunable three-dimensional engineered prostate cancer tissues for in vitro recapitulation of heterogeneous in vivo prostate tumor stiffness. Acta Biomater 2022; 147:73-90. [PMID: 35551999 DOI: 10.1016/j.actbio.2022.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022]
Abstract
In this manuscript we report the establishment and characterization of a three-dimensional in vitro, coculture engineered prostate cancer tissue (EPCaT) disease model based upon and informed by our characterization of in vivo prostate cancer (PCa) xenograft tumor stiffness. In prostate cancer, tissue stiffness is known to impact changes in gene and protein expression, alter therapeutic response, and be positively correlated with an aggressive clinical presentation. To inform an appropriate stiffness range for our in vitro model, PC-3 prostate tumor xenografts were established. Tissue stiffness ranged from 95 to 6,750 Pa. Notably, xenograft cell seeding density significantly impacted tumor stiffness; a two-fold increase in the number of seeded cells not only widened the tissue stiffness range throughout the tumor but also resulted in significant spatial heterogeneity. To fabricate our in vitro EPCaT model, PC-3 castration-resistant prostate cancer cells were co-encapsulated with BJ-5ta fibroblasts within a poly(ethylene glycol)-fibrinogen matrix augmented with excess poly(ethylene glycol)-diacrylate to modulate the matrix mechanical properties. Encapsulated cells temporally remodeled their in vitro microenvironment and enrichment of gene sets associated with tumorigenic progression was observed in response to increased matrix stiffness. Through variation of matrix composition and culture duration, EPCaTs were tuned to mimic the wide range of biomechanical cues provided to PCa cells in vivo; collectively, a range of 50 to 10,000 Pa was achievable. Markedly, this also encompasses published clinical PCa stiffness data. Overall, this study serves to introduce our bioinspired, tunable EPCaT model and provide the foundation for future PCa progression and drug development studies. STATEMENT OF SIGNIFICANCE: The development of cancer models that mimic the native tumor microenvironment (TME) complexities is critical to not only develop effective drugs but also enhance our understanding of disease progression. Here we establish and characterize our 3D in vitro engineered prostate cancer tissue model with tunable matrix stiffness, that is inspired by this study's spatial characterization of in vivo prostate tumor xenograft stiffness. Notably, our model's mimicry of the TME is further augmented by the inclusion of matrix remodeling fibroblasts to introduce cancer-stromal cell-cell interactions. This study addresses a critical unmet need in the field by elucidating the prostate tumor xenograft stiffness range and establishing a foundation for recapitulating the biomechanics of site-of-origin and soft tissue metastatic prostate tumors in vitro.
Collapse
Affiliation(s)
- Nicole L Habbit
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Benjamin Anbiah
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Luke Anderson
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Joshita Suresh
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Iman Hassani
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | - Matthew Eggert
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Andrew Brannen
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Joshua Davis
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Yuan Tian
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA
| | | | - Peter Panizzi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 362 Thach Concourse, Auburn, AL 36849, USA
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, 212 Ross Hall, Auburn, AL 36849, USA.
| |
Collapse
|
33
|
Zhang J, Tavakoli H, Ma L, Li X, Han L, Li X. Immunotherapy discovery on tumor organoid-on-a-chip platforms that recapitulate the tumor microenvironment. Adv Drug Deliv Rev 2022; 187:114365. [PMID: 35667465 DOI: 10.1016/j.addr.2022.114365] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/17/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has achieved remarkable success over the past decade by modulating patients' own immune systems and unleashing pre-existing immunity. However, only a minority of cancer patients across different cancer types are able to benefit from immunotherapy treatment; moreover, among those small portions of patients with response, intrinsic and acquired resistance remains a persistent challenge. Because the tumor microenvironment (TME) is well recognized to play a critical role in tumor initiation, progression, metastasis, and the suppression of the immune system and responses to immunotherapy, understanding the interactions between the TME and the immune system is a pivotal step in developing novel and efficient cancer immunotherapies. With unique features such as low reagent consumption, dynamic and precise fluid control, versatile structures and function designs, and 3D cell co-culture, microfluidic tumor organoid-on-a-chip platforms that recapitulate key factors of the TME and the immune contexture have emerged as innovative reliable tools to investigate how tumors regulate their TME to counteract antitumor immunity and the mechanism of tumor resistance to immunotherapy. In this comprehensive review, we focus on recent advances in tumor organoid-on-a-chip platforms for studying the interaction between the TME and the immune system. We first review different factors of the TME that recent microfluidic in vitro systems reproduce to generate advanced tools to imitate the crosstalk between the TME and the immune system. Then, we discuss their applications in the assessment of different immunotherapies' efficacy using tumor organoid-on-a-chip platforms. Finally, we present an overview and the outlook of engineered microfluidic platforms in investigating the interactions between cancer and immune systems, and the adoption of patient-on-a-chip models in clinical applications toward personalized immunotherapy.
Collapse
Affiliation(s)
- Jie Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China; Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Hamed Tavakoli
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Lei Ma
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Xiaochun Li
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Lichun Han
- Xi'an Daxing Hospital, Xi'an 710016, China
| | - XiuJun Li
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA; Border Biomedical Research Center, Forensic Science, & Environmental Science and Engineering, University of Texas at El Paso, 500 West University Ave., El Paso, TX 79968, USA.
| |
Collapse
|
34
|
Najibi AJ, Shih TY, Zhang DKY, Lou J, Sobral MC, Wang H, Dellacherie MO, Adu-Berchie K, Mooney DJ. Targeting tumor extracellular matrix activates the tumor-draining lymph nodes. Cancer Immunol Immunother 2022; 71:2957-2968. [PMID: 35524791 DOI: 10.1007/s00262-022-03212-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/15/2022] [Indexed: 11/26/2022]
Abstract
Disruption of the tumor extracellular matrix (ECM) may alter immune cell infiltration into the tumor and antitumor T cell priming in the tumor-draining lymph nodes (tdLNs). Here, we explore how intratumoral enzyme treatment (ET) of B16 melanoma tumors with ECM-depleting enzyme hyaluronidase alters adaptive and innate immune populations, including T cells, DCs, and macrophages, in the tumors and tdLNs. ET increased CD103+ DC abundance in the tdLNs, as well as antigen presentation of a model tumor antigen ovalbumin (OVA), eliciting local OVA-specific CD8+ T cell responses. Delivered in combination with a distant cryogel-based cancer vaccine, ET increased the systemic antigen-specific CD8+ T cell response. By enhancing activity within the tdLN, ET may broadly support immunotherapies in generating tumor-specific immunity.
Collapse
Affiliation(s)
- Alexander J Najibi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Ting-Yu Shih
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - David K Y Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Junzhe Lou
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Miguel C Sobral
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Hua Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Maxence O Dellacherie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Kwasi Adu-Berchie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Cameron AP, Zeng B, Liu Y, Wang H, Soheilmoghaddam F, Cooper-White J, Zhao CX. Biophysical properties of hydrogels for mimicking tumor extracellular matrix. BIOMATERIALS ADVANCES 2022; 136:212782. [PMID: 35929332 DOI: 10.1016/j.bioadv.2022.212782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/01/2022] [Accepted: 03/26/2022] [Indexed: 06/15/2023]
Abstract
The extracellular matrix (ECM) is an essential component of the tumor microenvironment. It plays a critical role in regulating cell-cell and cell-matrix interactions. However, there is lack of systematic and comparative studies on different widely-used ECM mimicking hydrogels and their properties, making the selection of suitable hydrogels for mimicking different in vivo conditions quite random. This study systematically evaluates the biophysical attributes of three widely used natural hydrogels (Matrigel, collagen gel and agarose gel) including complex modulus, loss tangent, diffusive permeability and pore size. A new and facile method was developed combining Critical Point Drying, Scanning Electron Microscopy imaging and a MATLAB image processing program (CSM method) for the characterization of hydrogel microstructures. This CSM method allows accurate measurement of the hydrogel pore size down to nanometer resolution. Furthermore, a microfluidic device was implemented to measure the hydrogel permeability (Pd) as a function of particle size and gel concentration. Among the three gels, collagen gel has the lowest complex modulus, medium pore size, and the highest loss tangent. Agarose gel exhibits the highest complex modulus, the lowest loss tangent and the smallest pore size. Collagen gel and Matrigel produced complex moduli close to that estimated for cancer ECM. The Pd of these hydrogels decreases significantly with the increase of particle size. By assessing different hydrogels' biophysical characteristics, this study provides valuable insights for tailoring their properties for various three-dimensional cancer models.
Collapse
Affiliation(s)
- Anna P Cameron
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Bijun Zeng
- Diamantina Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Yun Liu
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Haofei Wang
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Farhad Soheilmoghaddam
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Justin Cooper-White
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia; School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Chun-Xia Zhao
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
36
|
Qian X, Xu X, Wu Y, Wang J, Li J, Chen S, Wen J, Li Y, Zhang Z. Strategies of engineering nanomedicines for tumor retention. J Control Release 2022; 346:193-211. [PMID: 35447297 DOI: 10.1016/j.jconrel.2022.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 01/29/2023]
Abstract
The retention of therapeutic agents in solid tumors at sufficient concentration and duration is crucial for their antitumor effects. Given the important contribution of nanomedicines to oncology, we herein summarized two major strategies of nanomedicines for tumor retention, such as transformation- and interactions-mediated strategies. The transformation-mediated retention strategy was achieved by enlarging particle size of nanomedicines or modulating the morphology into fibrous structures, while the interactions-mediated retention strategy was accomplished by modulating nanomedicines to promote their interactions with versatile cells or components in tumors. Moreover, we provide some considerations and perspectives of tumor-retaining nanomedicines for effective cancer therapy.
Collapse
Affiliation(s)
- Xindi Qian
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Wu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shuo Chen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Jingyuan Wen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
37
|
Balancin ML, Baldavira CM, Prieto TG, Machado-Rugolo J, Farhat C, Assato AK, Velosa APP, Teodoro WR, Ab'Saber AM, Takagaki TY, Capelozzi VL. Dissecting and Reconstructing Matrix in Malignant Mesothelioma Through Histocell-Histochemistry Gradients for Clinical Applications. Front Med (Lausanne) 2022; 9:871202. [PMID: 35492318 PMCID: PMC9043486 DOI: 10.3389/fmed.2022.871202] [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: 02/07/2022] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMalignant pleural mesotheliomas (MM) are known for their heterogenous histology and clinical behavior. MM histology reveals three major tumor cell populations: epithelioid, sarcomatoid, and biphasic. Using a dissecting approach, we showed that histochemical gradients help us better understand tumor heterogeneity and reconsider its histologic classifications. We also showed that this method to characterize MM tumor cell populations provides a better understanding of the underlying mechanisms for invasion and disease progression.MethodsIn a cohort of 87 patients with surgically excised MM, we used hematoxylin and eosin to characterize tumor cell populations and Movat's pentachrome staining to dissect the ECM matrisome. Next, we developed a computerized semi-assisted protocol to quantify and reconstruct the ECM in 3D and examined the clinical association between the matricellular factors and patient outcome.ResultsEpithelioid cells had a higher matrix composition of elastin and fibrin, whereas, in the sarcomatoid type, hyaluronic acid and total collagen were most prevalent. The 3D reconstruction exposed the collagen I and III that form channels surrounding the neoplastic cell blocks. The estimated volume of the two collagen fractions was 14% of the total volume, consistent with the median estimated area of total collagen (12.05 mm2) for epithelioid MM.ConclusionDifferential patterns in matricellular phenotypes in MM could be used in translational studies to improve patient outcome. More importantly, our data raise the possibility that cancer cells can use the matrisome for disease expansion and could be effectively targeted by anti-collagen, anti-elastin, and/or anti-hyaluronic acid therapies.
Collapse
Affiliation(s)
- Marcelo Luiz Balancin
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Camila Machado Baldavira
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Tabatha Gutierrez Prieto
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Juliana Machado-Rugolo
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
- Health Technology Assessment Center (NATS), Clinical Hospital (HCFMB), Medical School of São Paulo State University (UNESP), Botucatu, Brazil
| | - Cecília Farhat
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Aline Kawassaki Assato
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Ana Paula Pereira Velosa
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, FMUSP, São Paulo, Brazil
| | - Walcy Rosolia Teodoro
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, FMUSP, São Paulo, Brazil
| | - Alexandre Muxfeldt Ab'Saber
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Teresa Yae Takagaki
- Division of Pneumology, Instituto do Coração (Incor), University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
- *Correspondence: Vera Luiza Capelozzi
| |
Collapse
|
38
|
Longitudinal shear wave elasticity measurements of millimeter-sized biomaterials using a single-element transducer platform. PLoS One 2022; 17:e0266235. [PMID: 35385536 PMCID: PMC8985960 DOI: 10.1371/journal.pone.0266235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/16/2022] [Indexed: 11/19/2022] Open
Abstract
Temporal variations of the extracellular matrix (ECM) stiffness profoundly impact cellular behaviors, possibly more significantly than the influence of static stiffness. Three-dimensional (3D) cell cultures with tunable matrix stiffness have been utilized to characterize the mechanobiological interactions of elasticity-mediated cellular behaviors. Conventional studies usually perform static interrogations of elasticity at micro-scale resolution. While such studies are essential for investigations of cellular mechanotransduction, few tools are available for depicting the temporal dynamics of the stiffness of the cellular environment, especially for optically turbid millimeter-sized biomaterials. We present a single-element transducer shear wave (SW) elasticity imaging system that is applied to a millimeter-sized, ECM-based cell-laden hydrogel. The single-element ultrasound transducer is used both to generate SWs and to detect their arrival times after being reflected from the side boundaries of the sample. The sample’s shear wave speed (SWS) is calculated by applying a time-of-flight algorithm to the reflected SWs. We use this noninvasive and technically straightforward approach to demonstrate that exposing 3D cancer cell cultures to X-ray irradiation induces a temporal change in the SWS. The proposed platform is appropriate for investigating in vitro how a group of cells remodels their surrounding matrix and how changes to their mechanical properties could affect the embedded cells in optically turbid millimeter-sized biomaterials.
Collapse
|
39
|
Esfahani P, Levine H, Mukherjee M, Sun B. Three-dimensional cancer cell migration directed by dual mechanochemical guidance. PHYSICAL REVIEW RESEARCH 2022; 4:L022007. [PMID: 37033157 PMCID: PMC10081505 DOI: 10.1103/physrevresearch.4.l022007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Directed cell migration guided by external cues plays a central role in many physiological and pathophysiological processes. The microenvironment of cells often simultaneously contains various cues and the motility response of cells to multiplexed guidance is poorly understood. Here we combine experiments and mathematical models to study the three-dimensional migration of breast cancer cells in the presence of both contact guidance and a chemoattractant gradient. We find that the chemotaxis of cells is complicated by the presence of contact guidance as the microstructure of extracellular matrix (ECM) vary spatially. In the presence of dual guidance, the impact of ECM alignment is determined externally by the coherence of ECM fibers and internally by cell mechanosensing Rho/Rock pathways. When contact guidance is parallel to the chemical gradient, coherent ECM fibers significantly increase the efficiency of chemotaxis. When contact guidance is perpendicular to the chemical gradient, cells exploit the ECM disorder to locate paths for chemotaxis. Our results underscore the importance of fully characterizing the cancer cell microenvironment in order to better understand invasion and metastasis.
Collapse
Affiliation(s)
- Pedram Esfahani
- Department of Physics, Oregon State University, Corvallis, Oregon 97331, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics, Northeastern University, Boston, Massachusetts 02115, USA
- Departments of Physics and Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| | - Mrinmoy Mukherjee
- Center for Theoretical Biological Physics, Northeastern University, Boston, Massachusetts 02115, USA
| | - Bo Sun
- Department of Physics, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
40
|
Biomimetic hydrogel supports initiation and growth of patient-derived breast tumor organoids. Nat Commun 2022; 13:1466. [PMID: 35304464 PMCID: PMC8933543 DOI: 10.1038/s41467-022-28788-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/01/2022] [Indexed: 12/15/2022] Open
Abstract
Patient-derived tumor organoids (PDOs) are a highly promising preclinical model that recapitulates the histology, gene expression, and drug response of the donor patient tumor. Currently, PDO culture relies on basement-membrane extract (BME), which suffers from batch-to-batch variability, the presence of xenogeneic compounds and residual growth factors, and poor control of mechanical properties. Additionally, for the development of new organoid lines from patient-derived xenografts, contamination of murine host cells poses a problem. We propose a nanofibrillar hydrogel (EKGel) for the initiation and growth of breast cancer PDOs. PDOs grown in EKGel have histopathologic features, gene expression, and drug response that are similar to those of their parental tumors and PDOs in BME. In addition, EKGel offers reduced batch-to-batch variability, a range of mechanical properties, and suppressed contamination from murine cells. These results show that EKGel is an improved alternative to BME matrices for the initiation, growth, and maintenance of breast cancer PDOs. Patient-derived tumour organoids are important preclinical models but suffer from variability from the use of basement-membrane extract and cell contamination. Here, the authors report on the development of mimetic nanofibrilar hydrogel which supports tumour organoid growth with reduced batch variability and cell contamination.
Collapse
|
41
|
G. K. AV, Gogoi G, Behera B, Rila S, Rangarajan A, Pandya HJ. RapidET: a MEMS-based platform for label-free and rapid demarcation of tumors from normal breast biopsy tissues. MICROSYSTEMS & NANOENGINEERING 2022; 8:1. [PMID: 35087680 PMCID: PMC8761751 DOI: 10.1038/s41378-021-00337-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/07/2021] [Accepted: 11/28/2021] [Indexed: 05/09/2023]
Abstract
The rapid and label-free diagnosis of malignancies in ex vivo breast biopsy tissues has significant utility in pathology laboratories and operating rooms. We report a MEMS-based platform integrated with microchips that performs phenotyping of breast biopsy tissues using electrothermal sensing. The microchip, fabricated on a silicon substrate, incorporates a platinum microheater, interdigitated electrodes (IDEs), and resistance temperature detectors (RTDs) as on-chip sensing elements. The microchips are integrated onto the platform using a slide-fit contact enabling quick replacement for biological measurements. The bulk resistivity (ρ B ), surface resistivity (ρ S ), and thermal conductivity (k) of deparaffinized and formalin-fixed paired tumor and adjacent normal breast biopsy samples from N = 8 patients were measured. For formalin-fixed samples, the mean ρ B for tumors showed a statistically significant fold change of 4.42 (P = 0.014) when the tissue was heated from 25 °C to 37 °C compared to the adjacent normal tissue, which showed a fold change of 3.47. The mean ρ S measurements also showed a similar trend. The mean k of the formalin-fixed tumor tissues was 0.309 ± 0.02 W m-1 K-1 compared to a significantly higher k of 0.563 ± 0.028 W m-1 K-1 for the adjacent normal tissues. A similar trend was observed in ρ B, ρ S, and k for the deparaffinized tissue samples. An analysis of a combination of ρ B , ρ S , and k using Fisher's combined probability test and linear regression suggests the advantage of using all three parameters simultaneously for distinguishing tumors from adjacent normal tissues with higher statistical significance.
Collapse
Affiliation(s)
- Anil Vishnu G. K.
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka India
| | - Gayatri Gogoi
- Department of Pathology, Assam Medical College, Dibrugarh, Assam India
| | - Bhagaban Behera
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, Karnataka India
| | - Saeed Rila
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, Karnataka India
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development, and Genetics, Indian Institute of Science, Bangalore, Karnataka India
| | - Hardik J. Pandya
- Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, Karnataka India
- Centre for Product Design and Manufacturing, Indian Institute of Science, Bangalore, Karnataka India
| |
Collapse
|
42
|
Prince E, Kheiri S, Wang Y, Xu F, Cruickshank J, Topolskaia V, Tao H, Young EWK, McGuigan AP, Cescon DW, Kumacheva E. Microfluidic Arrays of Breast Tumor Spheroids for Drug Screening and Personalized Cancer Therapies. Adv Healthc Mater 2022; 11:e2101085. [PMID: 34636180 DOI: 10.1002/adhm.202101085] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/30/2021] [Indexed: 12/20/2022]
Abstract
One of the obstacles limiting progress in the development of effective cancer therapies is the shortage of preclinical models that capture the dynamic nature of tumor microenvironments. Interstitial flow strongly impacts tumor response to chemotherapy; however, conventional in vitro cancer models largely disregard this key feature. Here, a proof of principle microfluidic platform for the generation of large arrays of breast tumor spheroids that are grown under close-to-physiological flow in a biomimetic hydrogel is reported. This cancer spheroids-on-a-chip model is used for time- and labor-efficient studies of the effects of drug dose and supply rate on the chemosensitivity of breast tumor spheroids. The capability to grow large arrays of tumor spheroids from patient-derived cells of different breast cancer subtypes is shown, and the correlation between in vivo drug efficacy and on-chip spheroid drug response is demonstrated. The proposed platform can serve as an in vitro preclinical model for the development of personalized cancer therapies and effective screening of new anticancer drugs.
Collapse
Affiliation(s)
- Elisabeth Prince
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Sina Kheiri
- Department of Mechanical & Industrial Engineering University of Toronto 5 King's College Circle Toronto Ontario M5S 3G8 Canada
| | - Yihe Wang
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Fei Xu
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Jennifer Cruickshank
- Princess Margaret Cancer Centre University Health Network 610 University Ave Toronto Ontario M5G 2C1 Canada
| | - Valentina Topolskaia
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Huachen Tao
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Edmond W. K. Young
- Department of Mechanical & Industrial Engineering University of Toronto 5 King's College Circle Toronto Ontario M5S 3G8 Canada
- Institute of Biomaterials and Biomedical Engineering University of Toronto 164 College St Toronto Ontario M5S 3G9 Canada
| | - Alison. P. McGuigan
- Institute of Biomaterials and Biomedical Engineering University of Toronto 164 College St Toronto Ontario M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry University of Toronto 200 College St Toronto Ontario M5S 3E5 Canada
| | - David W. Cescon
- Princess Margaret Cancer Centre University Health Network 610 University Ave Toronto Ontario M5G 2C1 Canada
| | - Eugenia Kumacheva
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
- Institute of Biomaterials and Biomedical Engineering University of Toronto 164 College St Toronto Ontario M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry University of Toronto 200 College St Toronto Ontario M5S 3E5 Canada
| |
Collapse
|
43
|
Xu X, Wu Y, Qian X, Wang Y, Wang J, Li J, Li Y, Zhang Z. Nanomedicine Strategies to Circumvent Intratumor Extracellular Matrix Barriers for Cancer Therapy. Adv Healthc Mater 2022; 11:e2101428. [PMID: 34706400 DOI: 10.1002/adhm.202101428] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/14/2021] [Indexed: 01/04/2023]
Abstract
The dense and heterogeneous physical network of the extracellular matrix (ECM) in tumors represents a formidable barrier that limits intratumor drug delivery and the therapeutic efficacy of many anticancer therapies. Here, the two major nanomedicine strategies to circumvent intratumor ECM barriers: regulating the physiochemical properties of nanomedicines and remodeling the components and structure of the ECM are summarized. Nanomedicines can be rationally regulated by optimizing physiochemical properties or designed with biomimetic features to promote ECM permeation capability. Meanwhile, they can also be designed to remodel the ECM by modulating signaling pathways or destroying the components and architecture of the ECM via chemical, biological, or physical treatments. These efforts produce profound improvements in intratumor drug delivery and anticancer efficacy. Moreover, to aid in their anticancer efficacy, feasible approaches for improving ECM-circumventing nanomedicines are proposed.
Collapse
Affiliation(s)
- Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- School of Pharmacy University of Chinese Academy of Sciences 19A Yuqian Road Beijing 100049 China
| | - Yao Wu
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
| | - Xindi Qian
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- School of Pharmacy University of Chinese Academy of Sciences 19A Yuqian Road Beijing 100049 China
| | - Yuqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- School of Pharmacy University of Chinese Academy of Sciences 19A Yuqian Road Beijing 100049 China
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai 201203 China
- School of Pharmacy University of Chinese Academy of Sciences 19A Yuqian Road Beijing 100049 China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations Yantai Institute of Materia Medica Shandong 264000 China
| |
Collapse
|
44
|
Shi Y, Sivarajan S, Xiang KM, Kostecki GM, Tung L, Crocker JC, Reich DH. Pervasive cytoquakes in the actomyosin cortex across cell types and substrate stiffness. Integr Biol (Camb) 2021; 13:246-257. [PMID: 34875067 DOI: 10.1093/intbio/zyab017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/14/2022]
Abstract
The actomyosin cytoskeleton enables cells to resist deformation, crawl, change their shape and sense their surroundings. Despite decades of study, how its molecular constituents can assemble together to form a network with the observed mechanics of cells remains poorly understood. Recently, it has been shown that the actomyosin cortex of quiescent cells can undergo frequent, abrupt reconfigurations and displacements, called cytoquakes. Notably, such fluctuations are not predicted by current physical models of actomyosin networks, and their prevalence across cell types and mechanical environments has not previously been studied. Using micropost array detectors, we have performed high-resolution measurements of the dynamic mechanical fluctuations of cells' actomyosin cortex and stress fiber networks. This reveals cortical dynamics dominated by cytoquakes-intermittent events with a fat-tailed distribution of displacements, sometimes spanning microposts separated by 4 μm, in all cell types studied. These included 3T3 fibroblasts, where cytoquakes persisted over substrate stiffnesses spanning the tissue-relevant range of 4.3 kPa-17 kPa, and primary neonatal rat cardiac fibroblasts and myofibroblasts, human embryonic kidney cells and human bone osteosarcoma epithelial (U2OS) cells, where cytoquakes were observed on substrates in the same stiffness range. Overall, these findings suggest that the cortex self-organizes into a marginally stable mechanical state whose physics may contribute to cell mechanical properties, active behavior and mechanosensing.
Collapse
Affiliation(s)
- Yu Shi
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Shankar Sivarajan
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Katherine M Xiang
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Geran M Kostecki
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - John C Crocker
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel H Reich
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
45
|
Labat B, Buchbinder N, Morin-Grognet S, Ladam G, Atmani H, Vannier JP. Biomimetic matrix for the study of neuroblastoma cells: A promising combination of stiffness and retinoic acid. Acta Biomater 2021; 135:383-392. [PMID: 34407473 DOI: 10.1016/j.actbio.2021.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023]
Abstract
Neuroblastoma is the third most common pediatric cancer composed of malignant immature cells that are usually treated pharmacologically by all trans-retinoic acid (ATRA) but sometimes, they can spontaneously differentiate into benign forms. In that context, biomimetic cell culture models are warranted tools as they can recapitulate many of the biochemical and biophysical cues of normal or pathological microenvironments. Inspired by that challenge, we developed a neuroblastoma culture system based on biomimetic LbL films of physiological biochemical composition and mechanical properties. For that, we used chondroitin sulfate A (CSA) and poly-L-lysine (PLL) that were assembled and mechanically tuned by crosslinking with genipin (GnP), a natural biocompatible crosslinker, in a relevant range of stiffness (30-160 kPa). We then assessed the adhesion, survival, motility, and differentiation of LAN-1 neuroblastoma cells. Remarkably, increasing the stiffness of the LbL films induced neuritogenesis that was strengthened by the combination with ATRA. These results highlight the crucial role of the mechanical cues of the neuroblastoma microenvironment since it can dramatically modulate the effect of pharmacologic drugs. In conclusion, our biomimetic platform offers a promising tool to help fundamental understanding and pharmacological screening of neuroblastoma differentiation and may assist the design of translational biomaterials to support neuronal regeneration. STATEMENT OF SIGNIFICANCE: Neuroblastoma is one of the most common pediatric tumor commonly treated by the administration of all-trans-retinoic acid (ATRA). Unfortunately, advanced neuroblastoma often develop ATRA resistance. Accordingly, in the field of pharmacological investigations on neuroblastoma, there is a tremendous need of physiologically relevant cell culture systems that can mimic normal or pathological extracellular matrices. In that context, we developed a promising matrix-like cell culture model that provides new insights on the crucial role of mechanical properties of the microenvironment upon the success of ATRA treatment on the neuroblastoma maturation. We were able to control adhesion, survival, motility, and differentiation of neuroblastoma cells. More broadly, we believe that our system will help the design of in vitro pharmacological screening strategy.
Collapse
Affiliation(s)
- Beatrice Labat
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France.
| | | | - Sandrine Morin-Grognet
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France
| | - Guy Ladam
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France
| | - Hassan Atmani
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France
| | - Jean-Pierre Vannier
- Normandie Univ, UNIROUEN, PANTHER - INSERM 1234 - UFR de Médecine et de Pharmacie de Rouen 22, boulevard Gambetta 76000 Rouen, France
| |
Collapse
|
46
|
Pavlova IP, Nair SS, Lundon D, Sobotka S, Roshandel R, Treacy PJ, Ratnani P, Brody R, Epstein JI, Ayala GE, Kyprianou N, Tewari AK. Multiphoton Microscopy for Identifying Collagen Signatures Associated with Biochemical Recurrence in Prostate Cancer Patients. J Pers Med 2021; 11:jpm11111061. [PMID: 34834413 PMCID: PMC8619628 DOI: 10.3390/jpm11111061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer is a heterogeneous disease that remains dormant for long periods or acts aggressively with poor clinical outcomes. Identifying aggressive prostate tumor behavior using current glandular-focused histopathological criteria is challenging. Recent evidence has implicated the stroma in modulating prostate tumor behavior and in predicting post-surgical outcomes. However, the emergence of stromal signatures has been limited, due in part to the lack of adoption of imaging modalities for stromal-specific profiling. Herein, label-free multiphoton microscopy (MPM), with its ability to image tissue with stromal-specific contrast, is used to identify prostate stromal features associated with aggressive tumor behavior and clinical outcome. MPM was performed on unstained prostatectomy specimens from 59 patients and on biopsy specimens from 17 patients with known post-surgery recurrence status. MPM-identified collagen content, organization, and morphological tumor signatures were extracted for each patient and screened for association with recurrent disease. Compared to tumors from patients whose disease did not recur, tumors from patients with recurrent disease exhibited higher MPM-identified collagen amount and collagen fiber intensity signal and width. Our study shows an association between MPM-identified stromal collagen features of prostate tumors and post-surgical disease recurrence, suggesting their potential for prostate cancer risk assessment.
Collapse
Affiliation(s)
- Ina P. Pavlova
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
- Correspondence: (I.P.P.); (A.K.T.); Tel.: +1-212-659-5654 (I.P.P.); +1-212-241-8711 (A.K.T.)
| | - Sujit S. Nair
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
| | - Dara Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
| | - Stanislaw Sobotka
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
| | - Reza Roshandel
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
| | | | - Parita Ratnani
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
| | - Rachel Brody
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Jonathan I. Epstein
- Department of Pathology, Urology and Oncology, Johns Hopkins Hospital, Baltimore, MD 21287, USA;
| | - Gustavo E. Ayala
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashutosh K. Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.S.N.); (D.L.); (S.S.); (R.R.); (P.R.); (N.K.)
- Correspondence: (I.P.P.); (A.K.T.); Tel.: +1-212-659-5654 (I.P.P.); +1-212-241-8711 (A.K.T.)
| |
Collapse
|
47
|
Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers (Basel) 2021; 13:3466. [PMID: 34298680 PMCID: PMC8303391 DOI: 10.3390/cancers13143466] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets. The recent appreciation of the key, complex and multiple roles of the ECM in cancer and of the CAF diversity, has revolutionized the field and raised innovative but challenging questions. Here, we rapidly present CAF heterogeneity in link with their specific ECM remodeling features observed in cancer, before developing each of the impacts of such ECM modifications on tumor progression (survival, angiogenesis, pre-metastatic niche, chemoresistance, etc.), and on patient prognosis. Finally, based on preclinical studies and recent results obtained from clinical trials, we highlight key mechanisms or proteins that are, or may be, used as potential therapeutic or diagnostic targets, and we report and discuss benefits, disappointments, or even failures, of recently reported stroma-targeting strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christine Jean
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31037 Toulouse, France; (I.B.); (S.Z.); (C.L.); (C.B.)
| |
Collapse
|
48
|
Drain AP, Zahir N, Northey JJ, Zhang H, Huang PJ, Maller O, Lakins JN, Yu X, Leight JL, Alston-Mills BP, Hwang ES, Chen YY, Park CC, Weaver VM. Matrix compliance permits NF-κB activation to drive therapy resistance in breast cancer. J Exp Med 2021; 218:e20191360. [PMID: 33822843 PMCID: PMC8025243 DOI: 10.1084/jem.20191360] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/07/2020] [Accepted: 02/12/2021] [Indexed: 01/10/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) are associated with poor survival mediated by treatment resistance. TNBCs are fibrotic, yet little is known regarding how the extracellular matrix (ECM) evolves following therapy and whether it impacts treatment response. Analysis revealed that while primary untreated TNBCs are surrounded by a rigid stromal microenvironment, chemotherapy-resistant residual tumors inhabit a softer niche. TNBC organoid cultures and xenograft studies showed that organoids interacting with soft ECM exhibit striking resistance to chemotherapy, ionizing radiation, and death receptor ligand TRAIL. A stiff ECM enhanced proapoptotic JNK activity to sensitize cells to treatment, whereas a soft ECM promoted treatment resistance by elevating NF-κB activity and compromising JNK activity. Treatment-resistant residual TNBCs residing within soft stroma had elevated activated NF-κB levels, and disengaging NF-κB activity sensitized tumors in a soft matrix to therapy. Thus, the biophysical properties of the ECM modify treatment response, and agents that modulate stiffness-dependent NF-κB or JNK activity could enhance therapeutic efficacy in patients with TNBC.
Collapse
Affiliation(s)
- Allison P. Drain
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
- University of California, Berkeley–University of California, San Francisco Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA
| | - Nastaran Zahir
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA
| | - Jason J. Northey
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Hui Zhang
- Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA
| | - Po-Jui Huang
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Ori Maller
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Johnathon N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Xinmiao Yu
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Jennifer L. Leight
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA
| | - Brenda P. Alston-Mills
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA
| | - E. Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Catherine C. Park
- Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA
- University of California, San Francisco Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
- University of California, San Francisco Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
49
|
Obermann WMJ, Brockhaus K, Eble JA. Platelets, Constant and Cooperative Companions of Sessile and Disseminating Tumor Cells, Crucially Contribute to the Tumor Microenvironment. Front Cell Dev Biol 2021; 9:674553. [PMID: 33937274 PMCID: PMC8085416 DOI: 10.3389/fcell.2021.674553] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Although platelets and the coagulation factors are components of the blood system, they become part of and contribute to the tumor microenvironment (TME) not only within a solid tumor mass, but also within a hematogenous micrometastasis on its way through the blood stream to the metastatic niche. The latter basically consists of blood-borne cancer cells which are in close association with platelets. At the site of the primary tumor, the blood components reach the TME via leaky blood vessels, whose permeability is increased by tumor-secreted growth factors, by incomplete angiogenic sprouts or by vasculogenic mimicry (VM) vessels. As a consequence, platelets reach the primary tumor via several cell adhesion molecules (CAMs). Moreover, clotting factor VII from the blood associates with tissue factor (TF) that is abundantly expressed on cancer cells. This extrinsic tenase complex turns on the coagulation cascade, which encompasses the activation of thrombin and conversion of soluble fibrinogen into insoluble fibrin. The presence of platelets and their release of growth factors, as well as fibrin deposition changes the TME of a solid tumor mass substantially, thereby promoting tumor progression. Disseminating cancer cells that circulate in the blood stream also recruit platelets, primarily by direct cell-cell interactions via different receptor-counterreceptor pairs and indirectly by fibrin, which bridges the two cell types via different integrin receptors. These tumor cell-platelet aggregates are hematogenous micrometastases, in which platelets and fibrin constitute a particular TME in favor of the cancer cells. Even at the distant site of settlement, the accompanying platelets help the tumor cell to attach and to grow into metastases. Understanding the close liaison of cancer cells with platelets and coagulation factors that change the TME during tumor progression and spreading will help to curb different steps of the metastatic cascade and may help to reduce tumor-induced thrombosis.
Collapse
Affiliation(s)
| | | | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
50
|
Drew J, Machesky LM. The liver metastatic niche: modelling the extracellular matrix in metastasis. Dis Model Mech 2021; 14:dmm048801. [PMID: 33973625 PMCID: PMC8077555 DOI: 10.1242/dmm.048801] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Dissemination of malignant cells from primary tumours to metastatic sites is a key step in cancer progression. Disseminated tumour cells preferentially settle in specific target organs, and the success of such metastases depends on dynamic interactions between cancer cells and the microenvironments they encounter at secondary sites. Two emerging concepts concerning the biology of metastasis are that organ-specific microenvironments influence the fate of disseminated cancer cells, and that cancer cell-extracellular matrix interactions have important roles at all stages of the metastatic cascade. The extracellular matrix is the complex and dynamic non-cellular component of tissues that provides a physical scaffold and conveys essential adhesive and paracrine signals for a tissue's function. Here, we focus on how extracellular matrix dynamics contribute to liver metastases - a common and deadly event. We discuss how matrix components of the healthy and premetastatic liver support early seeding of disseminated cancer cells, and how the matrix derived from both cancer and liver contributes to the changes in niche composition as metastasis progresses. We also highlight the technical developments that are providing new insights into the stochastic, dynamic and multifaceted roles of the liver extracellular matrix in permitting and sustaining metastasis. An understanding of the contribution of the extracellular matrix to different stages of metastasis may well pave the way to targeted and effective therapies against metastatic disease.
Collapse
Affiliation(s)
- James Drew
- CRUK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Laura M. Machesky
- CRUK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|