1
|
Devarasou S, Kang M, Shin JH. Biophysical perspectives to understanding cancer-associated fibroblasts. APL Bioeng 2024; 8:021507. [PMID: 38855445 PMCID: PMC11161195 DOI: 10.1063/5.0199024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024] Open
Abstract
The understanding of cancer has evolved significantly, with the tumor microenvironment (TME) now recognized as a critical factor influencing the onset and progression of the disease. This broader perspective challenges the traditional view that cancer is primarily caused by mutations, instead emphasizing the dynamic interaction between different cell types and physicochemical factors within the TME. Among these factors, cancer-associated fibroblasts (CAFs) command attention for their profound influence on tumor behavior and patient prognoses. Despite their recognized importance, the biophysical and mechanical interactions of CAFs within the TME remain elusive. This review examines the distinctive physical characteristics of CAFs, their morphological attributes, and mechanical interactions within the TME. We discuss the impact of mechanotransduction on CAF function and highlight how these cells communicate mechanically with neighboring cancer cells, thereby shaping the path of tumor development and progression. By concentrating on the biomechanical regulation of CAFs, this review aims to deepen our understanding of their role in the TME and to illuminate new biomechanical-based therapeutic strategies.
Collapse
Affiliation(s)
- Somayadineshraj Devarasou
- Department of Mechanical Engineering, Korea Advanced Institute of Science & Technology (KAIST), Daejeon, Korea
| | - Minwoo Kang
- Department of Mechanical Engineering, Korea Advanced Institute of Science & Technology (KAIST), Daejeon, Korea
| | - Jennifer H. Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science & Technology (KAIST), Daejeon, Korea
| |
Collapse
|
2
|
Ho NCW, Yap JYY, Zhao Z, Wang Y, Fernando K, Li CH, Kwang XL, Quah HS, Arcinas C, Iyer NG, Fong ELS. Bioengineered Hydrogels Recapitulate Fibroblast Heterogeneity in Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307129. [PMID: 38493497 PMCID: PMC11132030 DOI: 10.1002/advs.202307129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/30/2024] [Indexed: 03/19/2024]
Abstract
Recently mapped transcriptomic landscapes reveal the extent of heterogeneity in cancer-associated fibroblasts (CAFs) beyond previously established single-gene markers. Functional analyses of individual CAF subsets within the tumor microenvironment are critical to develop more accurate CAF-targeting therapeutic strategies. However, there is a lack of robust preclinical models that reflect this heterogeneity in vitro. In this study, single-cell RNA sequencing datasets acquired from head and neck squamous cell carcinoma tissues to predict microenvironmental and cellular features governing individual CAF subsets are leveraged. Some of these features are then incorporated into a tunable hyaluronan-based hydrogel system to culture patient-derived CAFs. Control over hydrogel degradability and integrin adhesiveness enabled derivation of the predominant myofibroblastic and inflammatory CAF subsets, as shown through changes in cell morphology and transcriptomic profiles. Last, using these hydrogel-cultured CAFs, microtubule dynamics are identified, but not actomyosin contractility, as a key mediator of CAF plasticity. The recapitulation of CAF heterogeneity in vitro using defined hydrogels presents unique opportunities for advancing the understanding of CAF biology and evaluation of CAF-targeting therapeutics.
Collapse
Affiliation(s)
- Nicholas Ching Wei Ho
- Translational Tumor Engineering Laboratory, Department of Biomedical EngineeringNational University of SingaporeSingapore119276Singapore
| | - Josephine Yu Yan Yap
- Translational Tumor Engineering Laboratory, Department of Biomedical EngineeringNational University of SingaporeSingapore119276Singapore
| | - Zixuan Zhao
- The N.1 Institute for HealthNational University of SingaporeSingapore117456Singapore
| | - Yunyun Wang
- Translational Tumor Engineering Laboratory, Department of Biomedical EngineeringNational University of SingaporeSingapore119276Singapore
| | - Kanishka Fernando
- Translational Tumor Engineering Laboratory, Department of Biomedical EngineeringNational University of SingaporeSingapore119276Singapore
| | - Constance H Li
- Cancer Therapeutics Research LaboratoryNational Cancer Centre SingaporeSingapore168583Singapore
- Duke‐NUS Medical SchoolNational University of SingaporeSingapore169857Singapore
| | - Xue Lin Kwang
- Cancer Therapeutics Research LaboratoryNational Cancer Centre SingaporeSingapore168583Singapore
| | - Hong Sheng Quah
- Cancer Therapeutics Research LaboratoryNational Cancer Centre SingaporeSingapore168583Singapore
- Duke‐NUS Medical SchoolNational University of SingaporeSingapore169857Singapore
| | - Camille Arcinas
- Duke‐NUS Medical SchoolNational University of SingaporeSingapore169857Singapore
| | - N. Gopalakrishna Iyer
- Cancer Therapeutics Research LaboratoryNational Cancer Centre SingaporeSingapore168583Singapore
- Duke‐NUS Medical SchoolNational University of SingaporeSingapore169857Singapore
| | - Eliza Li Shan Fong
- Translational Tumor Engineering Laboratory, Department of Biomedical EngineeringNational University of SingaporeSingapore119276Singapore
- The N.1 Institute for HealthNational University of SingaporeSingapore117456Singapore
- Cancer Science InstituteNational University of SingaporeSingapore117599Singapore
| |
Collapse
|
3
|
Liao Z, Lim JJH, Lee JXT, Chua D, Vos MIG, Yip YS, Too CB, Cao H, Wang JK, Shou Y, Tay A, Lehti K, Cheng HS, Tay CY, Tan NS. Attenuating Epithelial-to-Mesenchymal Transition in Cancer through Angiopoietin-Like 4 Inhibition in a 3D Tumor Microenvironment Model. Adv Healthc Mater 2024; 13:e2303481. [PMID: 37987244 DOI: 10.1002/adhm.202303481] [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: 10/11/2023] [Indexed: 11/22/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) plays a crucial role in metastatic cancer progression, and current research, which relies heavily on 2D monolayer cultures, falls short in recapitulating the complexity of a 3D tumor microenvironment. To address this limitation, a transcriptomic meta-analysis is conducted on diverse cancer types undergoing EMT in 2D and 3D cultures. It is found that mechanotransduction is elevated in 3D cultures and is further intensified during EMT, but not during 2D EMT. This analysis reveals a distinct 3D EMT gene signature, characterized by extracellular matrix remodeling coordinated by angiopoietin-like 4 (Angptl4) along with other canonical EMT regulators. Utilizing hydrogel-based 3D matrices with adjustable mechanical forces, 3D cancer cultures are established at varying physiological stiffness levels. A YAP:EGR-1 mediated up-regulation of Angptl4 expression is observed, accompanied by an upregulation of mesenchymal markers, at higher stiffness during cancer EMT. Suppression of Angptl4 using antisense oligonucleotides or anti-cAngptl4 antibodies leads to a dose-dependent abolishment of EMT-mediated chemoresistance and tumor self-organization in 3D, ultimately resulting in diminished metastatic potential and stunted growth of tumor xenografts. This unique programmable 3D cancer cultures simulate stiffness levels in the tumor microenvironment and unveil Angptl4 as a promising therapeutic target to inhibit EMT and impede cancer progression.
Collapse
Affiliation(s)
- Zehuan Liao
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore, 637551, Singapore
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Joseph Jing Heng Lim
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Jeannie Xue Ting Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Damien Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Marcus Ivan Gerard Vos
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Yun Sheng Yip
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Choon Boon Too
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Huan Cao
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Jun Kit Wang
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, N-7491, Norway
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Chor Yong Tay
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore, 637551, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| |
Collapse
|
4
|
Nguyen HD, Lin CC. Viscoelastic stiffening of gelatin hydrogels for dynamic culture of pancreatic cancer spheroids. Acta Biomater 2024; 177:203-215. [PMID: 38354874 PMCID: PMC10958777 DOI: 10.1016/j.actbio.2024.02.010] [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: 10/16/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
The tumor microenvironment (TME) in pancreatic adenocarcinoma (PDAC) is a complex milieu of cellular and non-cellular components. Pancreatic cancer cells (PCC) and cancer-associated fibroblasts (CAF) are two major cell types in PDAC TME, whereas the non-cellular components are enriched with extracellular matrices (ECM) that contribute to high stiffness and fast stress-relaxation. Previous studies have suggested that higher matrix rigidity promoted aggressive phenotypes of tumors, including PDAC. However, the effects of dynamic viscoelastic matrix properties on cancer cell fate remain largely unexplored. The focus of this work was to understand the effects of such dynamic matrix properties on PDAC cell behaviors, particularly in the context of PCC/CAF co-culture. To this end, we engineered gelatin-norbornene (GelNB) based hydrogels with a built-in mechanism for simultaneously increasing matrix elastic modulus and viscoelasticity. Two GelNB-based macromers, namely GelNB-hydroxyphenylacetic acid (GelNB-HPA) and GelNB-boronic acid (GelNB-BA), were modularly mixed and crosslinked with 4-arm poly(ethylene glycol)-thiol (PEG4SH) to form elastic hydrogels. Treating the hybrid hydrogels with tyrosinase not only increased the elastic moduli of the gels (due to HPA dimerization) but also concurrently produced 1,2-diols that formed reversible boronic acid-diol bonding with the BA groups on GelNB-BA. We employed patient-derived CAF and a PCC cell line COLO-357 to demonstrate the effect of increasing matrix stiffness and viscoelasticity on CAF and PCC cell fate. Our results indicated that in the stiffened environment, PCC underwent epithelial-mesenchymal transition. In the co-culture PCC and CAF spheroid, CAF enhanced PCC spreading and stimulated collagen 1 production. Through mRNA-sequencing, we further showed that stiffened matrices, regardless of the degree of stress-relaxation, heightened the malignant phenotype of PDAC cells. STATEMENT OF SIGNIFICANCE: The pancreatic cancer microenvironment is a complex milieu composed of various cell types and extracellular matrices. It has been suggested that stiffer matrices could promote aggressive behavior in pancreatic cancer, but the effect of dynamic stiffening and matrix stress-relaxation on cancer cell fate remains largely undefined. This study aimed to explore the impact of dynamic changes in matrix viscoelasticity on pancreatic ductal adenocarcinoma (PDAC) cell behavior by developing a hydrogel system capable of simultaneously increasing stiffness and stress-relaxation on demand. This is achieved by crosslinking two gelatin-based macromers through orthogonal thiol-norbornene photochemistry and post-gelation stiffening with mushroom tyrosinase. The results revealed that higher matrix stiffness, regardless of the degree of stress relaxation, exacerbated the malignant characteristics of PDAC cells.
Collapse
Affiliation(s)
- Han D Nguyen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
5
|
Micalet A, Pape J, Bakkalci D, Javanmardi Y, Hall C, Cheema U, Moeendarbary E. Evaluating the Impact of a Biomimetic Mechanical Environment on Cancer Invasion and Matrix Remodeling. Adv Healthc Mater 2023; 12:e2201749. [PMID: 36333907 PMCID: PMC11468596 DOI: 10.1002/adhm.202201749] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/10/2022] [Indexed: 10/13/2024]
Abstract
The stiffness of tumors and their host tissues is much higher than most hydrogels, which are conventionally used to study in vitro cancer progression. The tumoroid assay is an engineered 3D in vitro tumor model that allows investigation of cancer cell invasion in an environment that is biomimetic in terms of extracellular matrix (ECM) composition and stiffness. Using this model, the change in matrix stiffness by epithelial colorectal cancer cells is systematically characterized by atomic force microscopy indentation tests. Less invasive epithelial cancer cells stiffen the tumor microenvironment while highly aggressive epithelial cancer cells show significant softening of the tumor microenvironment. Changes in stiffness are attributed to both cell-generated active forces as well as ECM degradation and remodeling. The degradation is in part attributed to the enzymatic activity of matrix metalloproteinases (MMPs) as demonstrated by the significant expression of MMP-2 and MMP-9 at both gene and protein levels. Targeting MMP activity through broad-spectrum drug inhibition (BB-94) reverses the changes in stiffness and also decreases cancer cell invasion. These results promote the idea of using mechano-based cancer therapies such as MMP inhibition.
Collapse
Affiliation(s)
- Auxtine Micalet
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Judith Pape
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Deniz Bakkalci
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Yousef Javanmardi
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Chloe Hall
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Umber Cheema
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Emad Moeendarbary
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| |
Collapse
|
6
|
Devarasou S, Kang M, Kwon TY, Cho Y, Shin JH. Fibrous Matrix Architecture-Dependent Activation of Fibroblasts with a Cancer-Associated Fibroblast-like Phenotype. ACS Biomater Sci Eng 2023; 9:280-291. [PMID: 36573928 DOI: 10.1021/acsbiomaterials.2c00694] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are one of the most prevalent cell types within the tumor microenvironment (TME). While several physicochemical cues from the TME, including growth factors, cytokines, and ECM specificity, have been identified as essential factors for CAF activation, the precise mechanism of how the ECM architecture regulates CAF initiation remains elusive. Using a gelatin-based electrospun fiber mesh, we examined the effect of matrix fiber density on CAF activation induced by MCF-7 conditioned media (CM). A less dense (3D) gelatin mesh matrix facilitated better activation of dermal fibroblasts into a CAF-like phenotype in the CM than a highly dense (3D) gelatin mesh matrix. In addition, it was discovered that CAF activation on the less dense (LD) matrix is dependent on the cell size-related AKT/mTOR signaling cascade, accompanied by an increase in intracellular tension within the well-spread fibroblasts.
Collapse
Affiliation(s)
- Somayadineshraj Devarasou
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Minwoo Kang
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Tae Yoon Kwon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Youngbin Cho
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
7
|
Xu Y, Song D, Wang X. 3D Bioprinting for Pancreas Engineering/Manufacturing. Polymers (Basel) 2022; 14:polym14235143. [PMID: 36501537 PMCID: PMC9741443 DOI: 10.3390/polym14235143] [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: 10/11/2022] [Revised: 10/29/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetes is the most common chronic disease in the world, and it brings a heavy burden to people's health. Against this background, diabetic research, including islet functionalization has become a hot topic in medical institutions all over the world. Especially with the rapid development of microencapsulation and three-dimensional (3D) bioprinting technologies, organ engineering and manufacturing have become the main trends for disease modeling and drug screening. Especially the advanced 3D models of pancreatic islets have shown better physiological functions than monolayer cultures, suggesting their potential in elucidating the behaviors of cells under different growth environments. This review mainly summarizes the latest progress of islet capsules and 3D printed pancreatic organs and introduces the activities of islet cells in the constructs with different encapsulation technologies and polymeric materials, as well as the vascularization and blood glucose control capabilities of these constructs after implantation. The challenges and perspectives of the pancreatic organ engineering/manufacturing technologies have also been demonstrated.
Collapse
|
8
|
Hwang J, An EK, Zhang W, Kim HJ, Eom Y, Jin JO. Dual-functional alginate and collagen–based injectable hydrogel for the treatment of cancer and its metastasis. J Nanobiotechnology 2022; 20:245. [PMID: 35643505 PMCID: PMC9148466 DOI: 10.1186/s12951-022-01458-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/08/2022] [Indexed: 12/19/2022] Open
Abstract
Background Immunotherapies have been gaining attention for the prevention of cancer recurrence and metastasis. Cancer immunotherapy can induce memory cells to target cancer-specific antigens and, thus, selectively kill cancer cells. However, there are difficulties in inducing cancer antigen–specific immunity due to limited knowledge regarding cancer antigens. In this study, we synthesized a dual-functional hydrogel to induce antigen generation and immune activation. Results To elicit a cancer self-antigen–specific immune response, we synthesized an alginate-collagen–based injectable hydrogel, called thermally responsive hydrogel (pTRG), which was incorporated with indocyanine green and the immune stimulator polyinosinic:polycytidylic acid (poly I:C). pTRG was evaluated for its anticancer and anti-metastatic effects against CT-26 carcinoma and 4T1 breast tumor in mice by combining photothermal therapy (PTT) and immunotherapy. Near-infrared (NIR) irradiation promoted temperature elevation in pTRG, consequently exerting a therapeutic effect on mouse tumors. Lung metastasis was prevented in cured CT-26 tumor-injected mice following pTRG treatment via cancer antigen–specific T cell immunity. Moreover, pTRG successfully eliminated the original tumor in 4T1 tumor-bearing mice via PTT and protected them from lung metastasis. To further evaluate the carrier function of TRGs, different types of immunotherapeutic molecules were incorporated into TRGs, which led to the effective elimination of the first CT-26 tumor and the prevention of lung metastasis. Conclusions Our data demonstrate that TRG is a efficient material not only for treating primary tumors but also for preventing metastasis and recurrence.
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01458-x.
Collapse
|
9
|
Cao H, Yang L, Tian R, Wu H, Gu Z, Li Y. Versatile polyphenolic platforms in regulating cell biology. Chem Soc Rev 2022; 51:4175-4198. [PMID: 35535743 DOI: 10.1039/d1cs01165k] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polyphenolic materials are a class of fascinating and versatile bioinspired materials for biointerfacial engineering. In particular, due to the presence of active chemical groups, a series of unique physicochemical properties become accessible and tunable of the as-prepared polyphenolic platforms, which could delicately regulate the cell activities via cell-material contact-dependent interactions. More interestingly, polyphenols could also affect the cell behaviors via cell-material contact-independent manner, which arise due to their intrinsically functional characteristics (e.g., antioxidant and photothermal behaviors). As such, a comprehensive understanding on the relationship between material properties and desired biomedical applications, as well as the underlying mechanism at the cellular and molecular level would provide material design principles and accelerate the lab-to-clinic translation of polyphenolic platforms. In this review, we firstly give a brief overview of cell hallmarks governed by surrounding cues, followed by the introduction of polyphenolic material engineering strategies. Subsequently, a detailed discussion on cell-polyphenols contact-dependent interfacial interaction and contact-independent interaction was also carefully provided. Lastly, their biomedical applications were elaborated. We believe that this review could provide guidances for the rational material design of multifunctional polyphenols and extend their application window.
Collapse
Affiliation(s)
- Huan Cao
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Lei Yang
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Rong Tian
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Haoxing Wu
- Huaxi MR Research Center, Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhipeng Gu
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Yiwen Li
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Luo T, Tan B, Zhu L, Wang Y, Liao J. A Review on the Design of Hydrogels With Different Stiffness and Their Effects on Tissue Repair. Front Bioeng Biotechnol 2022; 10:817391. [PMID: 35145958 PMCID: PMC8822157 DOI: 10.3389/fbioe.2022.817391] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/07/2022] [Indexed: 12/20/2022] Open
Abstract
Tissue repair after trauma and infection has always been a difficult problem in regenerative medicine. Hydrogels have become one of the most important scaffolds for tissue engineering due to their biocompatibility, biodegradability and water solubility. Especially, the stiffness of hydrogels is a key factor, which influence the morphology of mesenchymal stem cells (MSCs) and their differentiation. The researches on this point are meaningful to the field of tissue engineering. Herein, this review focus on the design of hydrogels with different stiffness and their effects on the behavior of MSCs. In addition, the effect of hydrogel stiffness on the phenotype of macrophages is introduced, and then the relationship between the phenotype changes of macrophages on inflammatory response and tissue repair is discussed. Finally, the future application of hydrogels with a certain stiffness in regenerative medicine and tissue engineering has been prospected.
Collapse
Affiliation(s)
- Tianyi Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Bowen Tan
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lengjing Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yating Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jinfeng Liao,
| |
Collapse
|
11
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 294] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
12
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
13
|
Paradiso F, Serpelloni S, Francis LW, Taraballi F. Mechanical Studies of the Third Dimension in Cancer: From 2D to 3D Model. Int J Mol Sci 2021; 22:10098. [PMID: 34576261 PMCID: PMC8472581 DOI: 10.3390/ijms221810098] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/11/2022] Open
Abstract
From the development of self-aggregating, scaffold-free multicellular spheroids to the inclusion of scaffold systems, 3D models have progressively increased in complexity to better mimic native tissues. The inclusion of a third dimension in cancer models allows researchers to zoom out from a significant but limited cancer cell research approach to a wider investigation of the tumor microenvironment. This model can include multiple cell types and many elements from the extracellular matrix (ECM), which provides mechanical support for the tissue, mediates cell-microenvironment interactions, and plays a key role in cancer cell invasion. Both biochemical and biophysical signals from the extracellular space strongly influence cell fate, the epigenetic landscape, and gene expression. Specifically, a detailed mechanistic understanding of tumor cell-ECM interactions, especially during cancer invasion, is lacking. In this review, we focus on the latest achievements in the study of ECM biomechanics and mechanosensing in cancer on 3D scaffold-based and scaffold-free models, focusing on each platform's level of complexity, up-to-date mechanical tests performed, limitations, and potential for further improvements.
Collapse
Affiliation(s)
- Francesca Paradiso
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Stefano Serpelloni
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| | - Lewis W. Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea, Wales SA2 8PP, UK;
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA; (F.P.); (S.S.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| |
Collapse
|
14
|
Cao H, Cheng HS, Wang JK, Tan NS, Tay CY. A 3D physio-mimetic interpenetrating network-based platform to decode the pro and anti-tumorigenic properties of cancer-associated fibroblasts. Acta Biomater 2021; 132:448-460. [PMID: 33766799 DOI: 10.1016/j.actbio.2021.03.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/09/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Three-dimensional (3D) biomaterials with physiologically relevant and experimentally tractable biomechanical features are important platforms to advance our understanding of the influence of tissue mechanics in disease progression. Herein, an interpenetrating network (IPN) of collagen and alginate 3D culture system with tunable extracellular microstructure and mechanics is exploited as a tumor stroma proxy to study phenotypic plasticity of colorectal cancer-associated fibroblasts (CAF). In combination with Next Generation Sequencing (NGS) data analysis, we demonstrated that tuning the storage modulus of the IPN hydrogel between 49 and 419 Pa can trigger a reversible switch between an inflammatory (i-state, α-SMAlowIL-6high) and myofibroblastic (m-state, α-SMAhighIL-6low) state in CAF that is dependent on the polymer network confinement effect and ROS-HIF1-α mechanotransduction signaling axis. Secretome from m-state CAF upregulated several epithelial-mesenchymal-transition (EMT) transcripts and induced robust scattering in DLD-1, HCT116, and SW480 human colorectal adenocarcinoma, while the EMT-inducing capacity is muted in i-state CAF, suggestive of an anti-tumorigenic role. Our findings were further validated through Gene Expression Profiling Interactive Analysis (GEPIA), which showed that cytokines secreted at higher levels by i-state CAF are correlated (p < 0.05) with good overall colorectal cancer patient survival. Therefore, 3D network density and spatial cellular confinement are critical biophysical determinants that can profoundly influence CAF states, paracrine signaling, and EMT-inducing potential. STATEMENT OF SIGNIFICANCE: The communication between cancer cells and cancer-associated fibroblasts (CAF) contributes to tumor metastasis. CAF represent a diverse population of cellular subsets that can either promote or restrain tumor progression. However, the origin and cause of CAF heterogeneity remain elusive, limiting CAF-directed therapies for clinical use. We studied the dynamic phenotypes of CAF using a 3D physio-mimetic culture platform consisting of an interpenetrating collagen-alginate network. Combined with transcriptomic stratification and correlative analysis using cancer patient dataset, we showed phenotypic interconversion between inflammatory and myofibroblastic states, with anti- and pro-tumorigenic functions, in human colorectal CAF. This multidisciplinary study reveals the functional diversity of colorectal CAF caused by biophysical cues. The finding will influence the development of new CAF biomarkers and cancer therapies.
Collapse
|
15
|
Su C, Menon NV, Xu X, Teo YR, Cao H, Dalan R, Tay CY, Hou HW. A novel human arterial wall-on-a-chip to study endothelial inflammation and vascular smooth muscle cell migration in early atherosclerosis. LAB ON A CHIP 2021; 21:2359-2371. [PMID: 33978037 DOI: 10.1039/d1lc00131k] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Mechanistic understanding of atherosclerosis is largely hampered by the lack of a suitable in vitro human arterial model that recapitulates the arterial wall structure, and the interplay between different cell types and the surrounding extracellular matrix (ECM). This work introduces a novel microfluidic endothelial cell (EC)-smooth muscle cell (SMC) 3D co-culture platform that replicates the structural and biological aspects of the human arterial wall for modeling early atherosclerosis. Using a modified surface tension-based ECM patterning method, we established a well-defined intima-media-like structure, and identified an ECM composition (collagen I and Matrigel mixture) that retains the SMCs in a quiescent and aligned state, characteristic of a healthy artery. Endothelial stimulation with cytokines (IL-1β and TNFα) and oxidized low-density lipoprotein (oxLDL) was performed on-chip to study various early atherogenic events including endothelial inflammation (ICAM-1 expression), EC/SMC oxLDL uptake, SMC migration, and monocyte-EC adhesion. As a proof-of-concept for drug screening applications, we demonstrated the atheroprotective effects of vitamin D (1,25(OH)2D3) and metformin in mitigating cytokine-induced monocyte-EC adhesion and SMC migration. Overall, the developed arterial wall model facilitates quantitative and multi-factorial studies of EC and SMC phenotype in an atherogenic environment, and can be readily used as a platform technology to reconstitute multi-layered ECM tissue biointerfaces.
Collapse
Affiliation(s)
- Chengxun Su
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore. and Interdisciplinary Graduate School, Nanyang Technological University, Singapore, 639798, Singapore
| | - Nishanth Venugopal Menon
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore.
| | - Xiaohan Xu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore.
| | - Yu Rong Teo
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore.
| | - Huan Cao
- School of Materials Science & Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Rinkoo Dalan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore and Endocrinology Department, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - Chor Yong Tay
- School of Materials Science & Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore. and Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| |
Collapse
|
16
|
Micalet A, Moeendarbary E, Cheema U. 3D In Vitro Models for Investigating the Role of Stiffness in Cancer Invasion. ACS Biomater Sci Eng 2021. [PMID: 34081437 DOI: 10.1021/acsbiomaterials.0c01530] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tumorigenesis is attributed to the interactions of cancer cells with the tumor microenvironment through both biochemical cues and physical stimuli. Increased matrix deposition and realignment of the collagen fibers are detected by cancer cells, inducing epithelial-to-mesenchymal transition, which in turn stimulates cell motility and invasiveness. METHODS This review provides an overview of current research on the role of the physical microenvironment in cancer invasion. This was achieved by using a systematic approach and providing meta-analyses. Particular focus was placed on in vitro three-dimensional models of epithelial cancers. We investigated questions such as the effect of matrix stiffening, activation of stromal cells, and identified potential advances in mechano-based therapies. RESULTS Meta-analysis revealed that 64% of studies report cancer invasion promotion as stiffness increases, while 36% report the opposite. Experimental approaches and data interpretations were varied, each affecting the invasion of cancer differently. Examples are the experimental timeframes used (24 h to 21 days), the type of polymer used (24 types), and choice of cell line (33 cell lines). The stiffness of the 3D matrices varied from 0.5 to 300 kPa and 19% of these matrices' stiffness were outside commonly accepted physiological range. 100% of the studies outside biological stiffness range (above 20 kPa) report that stiffness does not promote cancer invasion. CONCLUSIONS Taking this analysis into account, we inform on the type of experimental approaches that could be the most relevant and provide what would be a standardized protocol and reporting strategy.
Collapse
Affiliation(s)
- Auxtine Micalet
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
| | - Umber Cheema
- Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| |
Collapse
|
17
|
Ort C, Chen Y, Ghagre A, Ehrlicher A, Moraes C. Bioprintable, Stiffness-Tunable Collagen-Alginate Microgels for Increased Throughput 3D Cell Culture Studies. ACS Biomater Sci Eng 2021; 7:2814-2822. [PMID: 34019377 DOI: 10.1021/acsbiomaterials.1c00129] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
3D culture platforms with tunable stiffness have the potential to improve many applications, such as drug discovery, organoid studies, and stem cell differentiation. Both dimensionality and stiffness regulate crucial and relevant cellular processes. However, 3D culture models are often limited in throughput and difficult to adopt for widespread use. Here, we demonstrate an accessible 3D, stiffness-tunable tissue culture platform, based on an interpenetrating network of collagen-1 and alginate. When blended with polymers that induce phase separation, these networks can be bioprinted at microliter volumes, using standard liquid handling infrastructure. We demonstrate robust reproducibility in printing these microgels, consistent tunability of mechanical properties, and maintained viability of multiple printed cell types. To highlight the utility and importance of this system, we demonstrate distinct morphological changes to cells in culture, use the system to probe the role of matrix mechanics and soluble factors in a collagen contraction assay, and perform a prototype viability screen against a candidate chemotherapeutic, demonstrating stiffness-dependent responses.
Collapse
Affiliation(s)
- Carley Ort
- Department of Chemical Engineering, McGill University, 3610 rue University, Montreal H3A 0G4, Quebec, Canada
| | - Yimai Chen
- Department of Chemical Engineering, McGill University, 3610 rue University, Montreal H3A 0G4, Quebec, Canada
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, 817 Sherbrooke Street West, Montreal H3A 2K6, Quebec, Canada
| | - Allen Ehrlicher
- Department of Biomedical Engineering, McGill University, 3775 rue University, Montreal H3A 2B4, Quebec, Canada.,Department of Bioengineering, McGill University, 817 Sherbrooke Street West, Montreal H3A 2K6, Quebec, Canada.,Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal H3A 1A3, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal H3A 0C7, Quebec, Canada.,Department of Mechanical Engineering, McGill University, Montreal H3A 0C3, Quebec, Canada
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University, 3610 rue University, Montreal H3A 0G4, Quebec, Canada.,Department of Biomedical Engineering, McGill University, 3775 rue University, Montreal H3A 2B4, Quebec, Canada.,Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal H3A 1A3, Quebec, Canada
| |
Collapse
|
18
|
Zhao Y, Zheng X, Zheng Y, Chen Y, Fei W, Wang F, Zheng C. Extracellular Matrix: Emerging Roles and Potential Therapeutic Targets for Breast Cancer. Front Oncol 2021; 11:650453. [PMID: 33968752 PMCID: PMC8100244 DOI: 10.3389/fonc.2021.650453] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence shows that the extracellular matrix (ECM) is an important regulator of breast cancer (BC). The ECM comprises of highly variable and dynamic components. Compared with normal breast tissue under homeostasis, the ECM undergoes many changes in composition and organization during BC progression. Induced ECM proteins, including fibrinogen, fibronectin, hyaluronic acid, and matricellular proteins, have been identified as important components of BC metastatic cells in recent years. These proteins play major roles in BC progression, invasion, and metastasis. Importantly, several specific ECM molecules, receptors, and remodeling enzymes are involved in promoting resistance to therapeutic intervention. Additional analysis of these ECM proteins and their downstream signaling pathways may reveal promising therapeutic targets against BC. These potential drug targets may be combined with new nanoparticle technologies. This review summarizes recent advances in functional nanoparticles that target the ECM to treat BC. Accurate nanomaterials may offer a new approach to BC treatment.
Collapse
Affiliation(s)
- Yunchun Zhao
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoling Zheng
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongquan Zheng
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fengmei Wang
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Lee HJ, Mun S, Pham DM, Kim P. Extracellular Matrix-Based Hydrogels to Tailoring Tumor Organoids. ACS Biomater Sci Eng 2021; 7:4128-4135. [DOI: 10.1021/acsbiomaterials.0c01801] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hyun Jin Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Siwon Mun
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Duc M. Pham
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
- Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| |
Collapse
|
20
|
Esmaeili J, Barati A, Ai J, Nooshabadi VT, Mirzaei Z. Employing hydrogels in tissue engineering approaches to boost conventional cancer-based research and therapies. RSC Adv 2021; 11:10646-10669. [PMID: 35423538 PMCID: PMC8695814 DOI: 10.1039/d1ra00855b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer is a complicated disease that involves the efforts of researchers to introduce and investigate novel successful treatments. Traditional cancer therapy approaches, especially chemotherapy, are prone to possible systemic side effects, such as the dysfunction of liver or kidney, neurological side effects and a decrease of bone marrow activity. Hydrogels, along with tissue engineering techniques, provide tremendous potential for scientists to overcome these issues through the release of drugs at the site of tumor. Hydrogels demonstrated competency as potent and stimulus-sensitive drug delivery systems for tumor removal, which is attributed to their unique features, including high water content, biocompatibility, and biodegradability. In addition, hydrogels have gained more attention as 3D models for easier and faster screening of cancer and tumors due to their potential in mimicking the extracellular matrix. Hydrogels as a reservoir can be loaded by an effective dosage of chemotherapeutic agents, and then deliver them to targets. In comparison to conventional procedures, hydrogels considerably decreased the total cost, duration of research, and treatment time. This study provides a general look into the potential role of hydrogels as a powerful tool to augment cancer studies for better analysis of cancerous cell functions, cell survival, angiogenesis, metastasis, and drug screening. Moreover, the upstanding application of drug delivery systems related to the hydrogel in order to sustain the release of desired drugs in the tumor cell-site were explored.
Collapse
Affiliation(s)
- Javad Esmaeili
- Department of Chemical Engineering, Faculty of Engineering, Arak University Arak Iran
- Department of Tissue Engineering, TISSUEHUB CO. Tehran Iran
| | - Abolfazl Barati
- Department of Chemical Engineering, Faculty of Engineering, Arak University Arak Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Technologies, Tehran University of Medical Sciences Tehran 14177-55469 Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Technologies, Tehran University of Medical Sciences Tehran 14177-55469 Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences Semnan Iran
| | - Zeynab Mirzaei
- Faculty of Biomedical Engineering, Amirkabir University of Technology Hafez str. 424 Tehran Iran
- Department of Tissue Engineering, TISSUEHUB CO. Tehran Iran
| |
Collapse
|
21
|
Yang J, Zhang Y, Qin M, Cheng W, Wang W, Cao Y. Understanding and Regulating Cell-Matrix Interactions Using Hydrogels of Designable Mechanical Properties. J Biomed Nanotechnol 2021; 17:149-168. [PMID: 33785089 DOI: 10.1166/jbn.2021.3026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Similar to natural tissues, hydrogels contain abundant water, so they are considered as promising biomaterials for studying the influence of the mechanical properties of extracellular matrices (ECM) on various cell functions. In recent years, the growing research on cellular mechanical response has revealed that many cell functions, including cell spreading, migration, tumorigenesis and differentiation, are related to the mechanical properties of ECM. Therefore, how cells sense and respond to the extracellular mechanical environment has gained considerable attention. In these studies, hydrogels are widely used as the in vitro model system. Hydrogels of tunable stiffness, viscoelasticity, degradability, plasticity, and dynamical properties have been engineered to reveal how cells respond to specific mechanical features. In this review, we summarize recent process in this research direction and specifically focus on the influence of the mechanical properties of the ECM on cell functions, how cells sense and respond to the extracellular mechanical environment, and approaches to adjusting the stiffness of hydrogels.
Collapse
Affiliation(s)
- Jiapeng Yang
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yu Zhang
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Meng Qin
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Wei Cheng
- Department of Oral Implantology Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Wei Wang
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yi Cao
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| |
Collapse
|
22
|
Blanco‐Fernandez B, Gaspar VM, Engel E, Mano JF. Proteinaceous Hydrogels for Bioengineering Advanced 3D Tumor Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003129. [PMID: 33643799 PMCID: PMC7887602 DOI: 10.1002/advs.202003129] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/13/2020] [Indexed: 05/14/2023]
Abstract
The establishment of tumor microenvironment using biomimetic in vitro models that recapitulate key tumor hallmarks including the tumor supporting extracellular matrix (ECM) is in high demand for accelerating the discovery and preclinical validation of more effective anticancer therapeutics. To date, ECM-mimetic hydrogels have been widely explored for 3D in vitro disease modeling owing to their bioactive properties that can be further adapted to the biochemical and biophysical properties of native tumors. Gathering on this momentum, herein the current landscape of intrinsically bioactive protein and peptide hydrogels that have been employed for 3D tumor modeling are discussed. Initially, the importance of recreating such microenvironment and the main considerations for generating ECM-mimetic 3D hydrogel in vitro tumor models are showcased. A comprehensive discussion focusing protein, peptide, or hybrid ECM-mimetic platforms employed for modeling cancer cells/stroma cross-talk and for the preclinical evaluation of candidate anticancer therapies is also provided. Further development of tumor-tunable, proteinaceous or peptide 3D microtesting platforms with microenvironment-specific biophysical and biomolecular cues will contribute to better mimic the in vivo scenario, and improve the predictability of preclinical screening of generalized or personalized therapeutics.
Collapse
Affiliation(s)
- Barbara Blanco‐Fernandez
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and TechnologyBaldiri Reixac 10–12Barcelona08028Spain
| | - Vítor M. Gaspar
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and TechnologyBaldiri Reixac 10–12Barcelona08028Spain
- Materials Science and Metallurgical EngineeringPolytechnical University of Catalonia (UPC)Eduard Maristany 16Barcelona08019Spain
- CIBER en BioingenieríaBiomateriales y NanomedicinaCIBER‐BBNMadrid28029Spain
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| |
Collapse
|
23
|
Fernando K, Kwang LG, Lim JTC, Fong ELS. Hydrogels to engineer tumor microenvironments in vitro. Biomater Sci 2021; 9:2362-2383. [DOI: 10.1039/d0bm01943g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Illustration of engineered hydrogel to recapitulate aspects of the tumor microenvironment.
Collapse
Affiliation(s)
- Kanishka Fernando
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
| | - Leng Gek Kwang
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
| | - Joanne Tze Chin Lim
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
| | - Eliza Li Shan Fong
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
- The N.1 Institute for Health
- National University of Singapore
| |
Collapse
|
24
|
Li Y, Khuu N, Prince E, Tao H, Zhang N, Chen Z, Gevorkian A, McGuigan AP, Kumacheva E. Matrix Stiffness-Regulated Growth of Breast Tumor Spheroids and Their Response to Chemotherapy. Biomacromolecules 2020; 22:419-429. [PMID: 33136364 DOI: 10.1021/acs.biomac.0c01287] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Interactions between tumor cells and the extracellular matrix (ECM) are an important factor contributing to therapy failure in cancer patients. Current in vitro breast cancer spheroid models examining the role of mechanical properties on spheroid response to chemotherapy are limited by the use of two-dimensional cell culture, as well as simultaneous variation in hydrogel matrix stiffness and other properties, e.g., hydrogel composition, pore size, and cell adhesion ligand density. In addition, currently used hydrogel matrices do not replicate the filamentous ECM architecture in a breast tumor microenvironment. Here, we report a collagen-alginate hydrogel with a filamentous architecture and a 20-fold variation in stiffness, achieved independently of other properties, used for the evaluation of estrogen receptor-positive breast cancer spheroid response to doxorubicin. The variation in hydrogel mechanical properties was achieved by altering the degree of cross-linking of alginate molecules. We show that soft hydrogels promote the growth of larger MCF-7 tumor spheroids with a lower fraction of proliferating cells and enhance spheroid resistance to doxorubicin. Notably, the stiffness-dependent chemotherapeutic response of the spheroids was temporally mediated: it became apparent at sufficiently long cell culture times, when the matrix stiffness has influenced the spheroid growth. These findings highlight the significance of decoupling matrix stiffness from other characteristics in studies of chemotherapeutic resistance of tumor spheroids and in development of drug screening platforms.
Collapse
Affiliation(s)
- Yunfeng Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China
| | - Nancy Khuu
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Elisabeth Prince
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Huachen Tao
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Ningtong Zhang
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Zhengkun Chen
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Albert Gevorkian
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Alison P McGuigan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E5, Canada.,The Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Eugenia Kumacheva
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E5, Canada.,The Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
25
|
Ort C, Lee W, Kalashnikov N, Moraes C. Disentangling the fibrous microenvironment: designer culture models for improved drug discovery. Expert Opin Drug Discov 2020; 16:159-171. [PMID: 32988224 DOI: 10.1080/17460441.2020.1822815] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Standard high-throughput screening (HTS) assays rarely identify clinically viable 'hits', likely because cells do not experience physiologically realistic culture conditions. The biophysical nature of the extracellular matrix has emerged as a critical driver of cell function and response and recreating these factors could be critically important in streamlining the drug discovery pipeline. AREAS COVERED The authors review recent design strategies to understand and manipulate biophysical features of three-dimensional fibrous tissues. The effects of architectural parameters of the extracellular matrix and their resulting mechanical behaviors are deconstructed; and their individual and combined impact on cell behavior is examined. The authors then illustrate the potential impact of these physical features on designing next-generation platforms to identify drugs effective against breast cancer. EXPERT OPINION Progression toward increased culture complexity must be balanced against the demanding technical requirements for high-throughput screening; and strategies to identify the minimal set of microenvironmental parameters needed to recreate disease-relevant responses must be specifically tailored to the disease stage and organ system being studied. Although challenging, this can be achieved through integrative and multidisciplinary technologies that span microfabrication, cell biology, and tissue engineering.
Collapse
Affiliation(s)
- Carley Ort
- Department of Chemical Engineering, McGill University , Montreal, Canada
| | - Wontae Lee
- Department of Chemical Engineering, McGill University , Montreal, Canada
| | - Nikita Kalashnikov
- Department of Chemical Engineering, McGill University , Montreal, Canada
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University , Montreal, Canada.,Department of Biomedical Engineering, McGill University , Montreal, Canada.,Rosalind & Morris Goodman Cancer Research Center, McGill University , Montreal, Canada
| |
Collapse
|
26
|
Wang JK, Cheam NMJ, Irvine SA, Tan NS, Venkatraman S, Tay CY. Interpenetrating Network of Alginate–Human Adipose Extracellular Matrix Hydrogel for Islet Cells Encapsulation. Macromol Rapid Commun 2020; 41:e2000275. [DOI: 10.1002/marc.202000275] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/04/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Jun Kit Wang
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
| | - Nicole Mein Ji Cheam
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
| | - Scott Alexander Irvine
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
| | - Nguan Soon Tan
- School of Biological Sciences Nanyang Technological University Singapore 60 Nanyang Drive Singapore 637551 Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore 11 Mandalay Road Singapore 308232 Singapore
| | - Subbu Venkatraman
- Department of Materials Science and Engineering National University of Singapore Blk EA, 9 Engineering Drive 1 Singapore 117575 Singapore
| | - Chor Yong Tay
- School of Materials Science and Engineering Nanyang Technological University Singapore N4.1, 50 Nanyang Avenue Singapore 639798 Singapore
- School of Biological Sciences Nanyang Technological University Singapore 60 Nanyang Drive Singapore 637551 Singapore
- Environmental Chemistry and Materials Centre Nanyang Environment and Water Research Institute 1 CleanTech Loop, CleanTech One Singapore 637141 Singapore
| |
Collapse
|
27
|
Wang M, Yang Y, Han L, Han S, Liu N, Xu F, Li F. Effect of three-dimensional ECM stiffness on cancer cell migration through regulating cell volume homeostasis. Biochem Biophys Res Commun 2020; 528:459-465. [PMID: 32505356 DOI: 10.1016/j.bbrc.2020.05.182] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022]
Abstract
The extracellular matrix (ECM) stiffness has direct effect on cancer cells homeostasis (e.g., cell volume), which is critical for regulation of their migration. However, the relationship among ECM stiffness, cell volume and cancer cell migration in three-dimensional (3D) microenvironment remains elusive. In this work, we prepared the collagen-alginate hydrogels with tunable stiffness to study how the 3D ECM stiffness influences cell volume and their migration. We found the cell volume homeostasis and migration speed of the MDA-MB-231 cells are both regulated by 3D ECM stiffness, while cell migration speed shows the same stiffness-dependent trend with cell volume. Deviating the cell volume from its homeostasis state can cause a significant decrease in its migration ability, which can be recovered through recovering the cell volume to its homeostasis state. This work reveals for the first time that 3D ECM stiffness regulates cell migration behavior through regulating cell volume homeostasis, which may provide a novel view in the exploration of the underlying mechanisms of cancer metastasis and cellular mechanotransduction.
Collapse
Affiliation(s)
- Meng Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Yaowei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Lichun Han
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China; Department of Anesthesia, Xi'an Daxing Hospital, Xi'an, 710049, PR China
| | - Shuang Han
- Department of Digestive Diseases, Hong Hui Hospital, Xi'an, 710054, PR China
| | - Na Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Fei Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China.
| |
Collapse
|
28
|
Takahara A. Preface to the Interfaces and Biology 1: Mechanobiology Special Issue. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7333-7334. [PMID: 31181916 DOI: 10.1021/acs.langmuir.9b01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
|