1
|
Andersen MK, Krossa S, Midtbust E, Pedersen CA, Wess M, Høiem TS, Viset T, Størkersen Ø, Nervik I, Sandsmark E, Bertilsson H, Giskeødegård GF, Rye MB, Tessem MB. Spatial transcriptomics reveals strong association between SFRP4 and extracellular matrix remodeling in prostate cancer. Commun Biol 2024; 7:1462. [PMID: 39511287 PMCID: PMC11543834 DOI: 10.1038/s42003-024-07161-x] [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: 05/23/2023] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
Prostate tumor heterogeneity is a major obstacle when studying the biological mechanisms of molecular markers. Increased gene expression levels of secreted frizzled-related protein 4 (SFRP4) is a biomarker in aggressive prostate cancer. To understand how SFRP4 relates to prostate cancer we performed comprehensive spatial and multiomics analysis of the same prostate cancer tissue samples. The experimental workflow included spatial transcriptomics, bulk transcriptomics, proteomics, DNA methylomics and tissue staining. SFRP4 mRNA was predominantly located in cancer stroma, produced by fibroblasts and smooth muscle cells, and co-expressed with extracellular matrix components. We also confirmed that higher SFRP4 gene expression is associated with cancer aggressiveness. Gene expression of SFRP4 was affected by gene promotor methylation. Surprisingly, the high mRNA levels did not reflect SFRP4 protein levels, which was much lower. This study contributes previously unknown insights of SFRP4 mRNA in the prostate tumor environment that potentially can improve diagnosis and treatment.
Collapse
Affiliation(s)
- Maria K Andersen
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Sebastian Krossa
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Central staff, St. Olavs Hospital HF, Trondheim, Norway
| | - Elise Midtbust
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Christine A Pedersen
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Maximilian Wess
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Therese S Høiem
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Trond Viset
- Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Øystein Størkersen
- Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ingunn Nervik
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Elise Sandsmark
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Helena Bertilsson
- Central staff, St. Olavs Hospital HF, Trondheim, Norway
- Central Norway Regional Health Authority, Stjørdal, Norway
| | - Guro F Giskeødegård
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Morten B Rye
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St.Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - May-Britt Tessem
- Department of Circulation and Medical Imaging, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
2
|
Yu H, Yan Z, Dreiss CA, Gaitano GG, Jarvis JA, Gentleman E, da Silva RMP, Grigoriadis AE. Injectable PEG Hydrogels with Tissue-Like Viscoelasticity Formed through Reversible Alendronate-Calcium Phosphate Crosslinking for Cell-Material Interactions. Adv Healthc Mater 2024; 13:e2400472. [PMID: 38809180 DOI: 10.1002/adhm.202400472] [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: 02/06/2024] [Indexed: 05/30/2024]
Abstract
Synthetic hydrogels provide controllable 3D environments, which can be used to study fundamental biological phenomena. The growing body of evidence that cell behavior depends upon hydrogel stress relaxation creates a high demand for hydrogels with tissue-like viscoelastic properties. Here, a unique platform of synthetic polyethylene glycol (PEG) hydrogels in which star-shaped PEG molecules are conjugated with alendronate and/or RGD peptides, attaining modifiable degradability as well as flexible cell adhesion, is created. Novel reversible ionic interactions between alendronate and calcium phosphate nanoparticles, leading to versatile viscoelastic properties with varying initial elastic modulus and stress relaxation time, are identified. This new crosslinking mechanism provides shear-thinning properties resulting in differential cellular responses between cancer cells and stem cells. The novel hydrogel system is an improved design to the other ionic crosslink platforms and opens new avenues for the development of pathologically relevant cancer models, as well as minimally invasive approaches for cell delivery for potential regenerative therapies.
Collapse
Affiliation(s)
- Hongqiang Yu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Ziqian Yan
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Cecile A Dreiss
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Gustavo G Gaitano
- Department of Chemistry, University of Navarra, Pamplona, 31080, Spain
| | - James A Jarvis
- Randall Division of Cell and Molecular Biophysics and NMR Facility, Centre for Biomolecular Spectroscopy, King's College London, London, SE1 1UL, UK
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Ricardo M P da Silva
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | | |
Collapse
|
3
|
Dong G, Wang J, Chen Z, Wang F, Xia B, Chen G. Regulatory effects of stress release from decellularized periosteum on proliferation, migration, and osteogenic differentiation of periosteum-derived cells. Biomater Sci 2024; 12:3360-3373. [PMID: 38771565 DOI: 10.1039/d4bm00358f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Bone injury is often associated with tears in the periosteum and changes in the internal stress microenvironment of the periosteum. In this study, we investigated the biological effects of periosteal prestress release on periosteum-derived cells (PDCs) and the potential mechanisms of endogenous stem cell recruitment. Decellularized periosteum with natural extracellular matrix (ECM) components was obtained by a combination of physical, chemical, and enzymatic decellularization. The decellularized periosteum removed immunogenicity while retaining the natural network structure and composition of the ECM. The Young's modulus has no significant difference between the periosteum before and after decellularization. The extracted PDCs were further composited with the decellularized periosteum and subjected to 20% stress release. It was found that the proliferative capacity of PDCs seeded on decellularized periosteum was significantly enhanced 6 h after stress release of the periosteum. The cell culture supernatant obtained after periosteal prestress release was able to significantly promote the migration ability of PDCs within 24 h. Enzyme-linked immunosorbnent assay (ELISA) experiments showed that the expression of stroma-derived factor-1α (SDF-1α) and vascular endothelial growth factor (VEGF) in the supernatant increased significantly after 3 h and 12 h of stress release, respectively. Furthermore, periosteal stress release promoted the high expression of osteogenic markers osteocalcin (OCN), osteopontin (OPN), and collagen type I of PDCs. The change in stress environment caused by the release of periosteal prestress was sensed by integrin β1, a mechanoreceptor on the membrane of PDCs, which further stimulated the expression of YAP in the nucleus. These investigations provided a novel method to evaluate the importance of mechanical stimulation in periosteum, which is also of great significance for the design and fabrication of artificial periosteum with mechanical regulation function.
Collapse
Affiliation(s)
- Gangli Dong
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Jinsong Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Zhongmin Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Fuping Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Bin Xia
- Engineering Research Center for Waste Oil Recovery Technology and Equipment, Ministry of Education, Chongqing Technology and Business University, Chongqing 400067, P. R. China.
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| |
Collapse
|
4
|
Guimarães CF, Liu S, Wang J, Purcell E, Ozedirne T, Ren T, Aslan M, Yin Q, Reis RL, Stoyanova T, Demirci U. Co-axial hydrogel spinning for facile biofabrication of prostate cancer-like 3D models. Biofabrication 2024; 16:025017. [PMID: 38306674 DOI: 10.1088/1758-5090/ad2535] [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: 06/09/2023] [Accepted: 02/01/2024] [Indexed: 02/04/2024]
Abstract
Glandular cancers are amongst the most prevalent types of cancer, which can develop in many different organs, presenting challenges in their detection as well as high treatment variability and failure rates. For that purpose, anticancer drugs are commonly tested in cancer cell lines grown in 2D tissue culture on plastic dishesin vitro, or in animal modelsin vivo. However, 2D culture models diverge significantly from the 3D characteristics of living tissues and animal models require extensive animal use and time. Glandular cancers, such as prostate cancer-the second leading cause of male cancer death-typically exist in co-centrical architectures where a cell layer surrounds an acellular lumen. Herein, this spatial cellular position and 3D architecture, containing dual compartments with different hydrogel materials, is engineered using a simple co-axial nozzle setup, in a single step utilizing prostate as a model of glandular cancer. The resulting hydrogel soft structures support viable prostate cancer cells of different cell lines and enable over-time maturation into cancer-mimicking aggregates surrounding the acellular core. The biofabricated cancer mimicking structures are then used as a model to predict the inhibitory efficacy of the poly ADP ribose polymerase inhibitor, Talazoparib, and the antiandrogen drug, Enzalutamide, in the growth of the cancer cell layer. Our results show that the obtained hydrogel constructs can be adapted to quickly obtain 3D cancer models which combine 3D physiological architectures with high-throughput screening to detect and optimize anti-cancer drugs in prostate and potentially other glandular cancer types.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga and Guimarães, Portugal
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Shiqin Liu
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States of America
| | - Jie Wang
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Emma Purcell
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Tugba Ozedirne
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Tanchen Ren
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Merve Aslan
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Qingqing Yin
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| | - Rui L Reis
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - Tanya Stoyanova
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States of America
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States of America
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Bio-Acoustic MEMS (BAMM) in Medicine Lab, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
- Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, United States of America
| |
Collapse
|
5
|
Zhou C, Yang Y, Duan M, Chen C, Pi C, Zhang D, Liu X, Xie J. Biomimetic Fibers Based on Equidistant Micropillar Arrays Determines Chondrocyte Fate via Mechanoadaptability. Adv Healthc Mater 2023; 12:e2301685. [PMID: 37596884 DOI: 10.1002/adhm.202301685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/02/2023] [Indexed: 08/20/2023]
Abstract
It is recognized that the changes in the physical properties of extracellular matrix (ECM) result in fine-tuned cell responses including cell morphology, proliferation and differentiation. In this study, a novel patterned equidistant micropillar substrate based on polydimethylsiloxane (PDMS) is designed to mimic the collagen fiber-like network of the cartilage matrix. By changing the component of the curing agent to an oligomeric base, micropillar substrates with the same topology but different stiffnesses are obtained and it is found that chondrocytes seeded onto the soft micropillar substrate maintain their phenotype by gathering type II collagen and aggrecan more effectively than those seeded onto the stiff micropillar substrate. Moreover, chondrocytes sense and respond to micropillar substrates with different stiffnesses by altering the ECM-cytoskeleton-focal adhesion axis. Further, it is found that the soft substrate-preserved chondrocyte phenotype is dependent on the activation of Wnt/β-catenin signaling. Finally, it is indicated that the changes in osteoid-like region formation and cartilage phenotype loss in the stiffened sclerotic area of osteoarthritis cartilage to validate the changes triggered by micropillar substrates with different stiffnesses. This study provides the cell behavior changes that are more similar to those of real chondrocytes at tissue level during the transition from a normal state to a state of osteoarthritis.
Collapse
Affiliation(s)
- Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Yueyi Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610064, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610064, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
6
|
Liu DM, Wu ZX, Guan JY. Intercellular competitive growth dynamics with microenvironmental feedback. Phys Rev E 2023; 108:054105. [PMID: 38115538 DOI: 10.1103/physreve.108.054105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Normal life activities between cells rely crucially on the homeostasis of the cellular microenvironment, but aging and cancer will upset this balance. In this paper we introduce the microenvironmental feedback mechanism to the growth dynamics of multicellular organisms, which changes the cellular competitive ability and thereby regulates the growth of multicellular organisms. We show that the presence of microenvironmental feedback can effectively delay aging, but cancer cells may grow uncontrollably due to the emergence of the tumor microenvironment (TME). We study the effect of the fraction of cancer cells relative to that of senescent cells on the feedback rate of the microenvironment on the lifespan of multicellular organisms and find that the average lifespan shortened is close to the data for non-Hodgkin's lymphoma in Canada from 1980 to 2015. We also investigate how the competitive ability of cancer cells affects the lifespan of multicellular organisms and reveal that there is an optimal value of the competitive ability of cancer cells allowing the organism to survive longest. Interestingly, the proposed microenvironmental feedback mechanism can give rise to the phenomenon of Parrondo's paradox: When the competitive ability of cancer cells switches between a too-high and a too-low value, multicellular organisms are able to live longer than in each case individually. Our results may provide helpful clues for targeted therapies aimed at the TME.
Collapse
Affiliation(s)
- De-Ming Liu
- Lanzhou Center for Theoretical Physics, Key Laboratory of Theoretical Physics of Gansu Province, and Key Laboratory of Quantum Theory and Applications of MoE, Lanzhou University, Lanzhou, Gansu 730000, China and Institute of Computational Physics and Complex Systems, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zhi-Xi Wu
- Lanzhou Center for Theoretical Physics, Key Laboratory of Theoretical Physics of Gansu Province, and Key Laboratory of Quantum Theory and Applications of MoE, Lanzhou University, Lanzhou, Gansu 730000, China and Institute of Computational Physics and Complex Systems, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jian-Yue Guan
- Lanzhou Center for Theoretical Physics, Key Laboratory of Theoretical Physics of Gansu Province, and Key Laboratory of Quantum Theory and Applications of MoE, Lanzhou University, Lanzhou, Gansu 730000, China and Institute of Computational Physics and Complex Systems, Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
7
|
Martino F, Lupi M, Giraudo E, Lanzetti L. Breast cancers as ecosystems: a metabolic perspective. Cell Mol Life Sci 2023; 80:244. [PMID: 37561190 PMCID: PMC10415483 DOI: 10.1007/s00018-023-04902-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer and one of the major causes of cancer death. Despite enormous progress in its management, both from the therapeutic and early diagnosis viewpoints, still around 700,000 patients succumb to the disease each year, worldwide. Late recurrency is the major problem in BC, with many patients developing distant metastases several years after the successful eradication of the primary tumor. This is linked to the phenomenon of metastatic dormancy, a still mysterious trait of the natural history of BC, and of several other types of cancer, by which metastatic cells remain dormant for long periods of time before becoming reactivated to initiate the clinical metastatic disease. In recent years, it has become clear that cancers are best understood if studied as ecosystems in which the impact of non-cancer-cell-autonomous events-dependent on complex interaction between the cancer and its environment, both local and systemic-plays a paramount role, probably as significant as the cell-autonomous alterations occurring in the cancer cell. In adopting this perspective, a metabolic vision of the cancer ecosystem is bound to improve our understanding of the natural history of cancer, across space and time. In BC, many metabolic pathways are coopted into the cancer ecosystem, to serve the anabolic and energy demands of the cancer. Their study is shedding new light on the most critical aspect of BC management, of metastatic dissemination, and that of the related phenomenon of dormancy and fostering the application of the knowledge to the development of metabolic therapies.
Collapse
Affiliation(s)
- Flavia Martino
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Mariadomenica Lupi
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Enrico Giraudo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Science and Drug Technology, University of Torino, Turin, Italy
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Turin, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
| |
Collapse
|
8
|
Papalazarou V, Drew J, Juin A, Spence HJ, Whitelaw J, Nixon C, Salmeron-Sanchez M, Machesky LM. Collagen VI expression is negatively mechanosensitive in pancreatic cancer cells and supports the metastatic niche. J Cell Sci 2022; 135:jcs259978. [PMID: 36546396 PMCID: PMC9845737 DOI: 10.1242/jcs.259978] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 11/19/2022] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is a deadly and highly metastatic disease, although how metastatic lesions establish is not fully understood. A key feature of pancreatic tumours is extensive fibrosis and deposition of extracellular matrix (ECM). While pancreatic cancer cells are programmed by stimuli derived from a stiff ECM, metastasis requires loss of attachment and adaptation to a softer microenvironment at distant sites. Growing evidence suggests that stiff ECM influences pancreatic cancer cell behaviour. Here, we argue that this influence is reversible and that pancreatic cancer cells can be reprogrammed upon sensing soft substrates. Using engineered polyacrylamide hydrogels with tuneable mechanical properties, we show that collagen VI is specifically upregulated in pancreatic cancer cells on soft substrates, due to a lack of integrin engagement. Furthermore, the expression of collagen VI is inversely correlated with mechanosensing and activity of YAP (also known as YAP1), which might be due to a direct or indirect effect on transcription of genes encoding collagen VI. Collagen VI supports migration in vitro and metastasis formation in vivo. Metastatic nodules formed by pancreatic cancer cells lacking Col6a1 display stromal cell-derived collagen VI deposition, suggesting that collagen VI derived from either cancer cells or the stroma is an essential component of the metastatic niche. This article has an associated First Person interview with Vasileios Papalazarou, joint first author of the paper.
Collapse
Affiliation(s)
- Vasileios Papalazarou
- Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Centre for the Cellular Microenvironment, University of Glasgow,Glasgow G11 6EW, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - James Drew
- Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Amelie Juin
- Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Heather J. Spence
- Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Jamie Whitelaw
- Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Colin Nixon
- Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | | | - Laura M. Machesky
- Cancer Research UK Beatson Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| |
Collapse
|
9
|
Najmina M, Ebara M, Ohmura T, Uto K. Viscoelastic Liquid Matrix with Faster Bulk Relaxation Time Reinforces the Cell Cycle Arrest Induction of the Breast Cancer Cells via Oxidative Stress. Int J Mol Sci 2022; 23:ijms232314637. [PMID: 36498966 PMCID: PMC9736955 DOI: 10.3390/ijms232314637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
The reactivating of disseminated dormant breast cancer cells in a soft viscoelastic matrix is mostly correlated with metastasis. Metastasis occurs due to rapid stress relaxation owing to matrix remodeling. Here, we demonstrate the possibility of promoting the permanent cell cycle arrest of breast cancer cells on a viscoelastic liquid substrate. By controlling the molecular weight of the hydrophobic molten polymer, poly(ε-caprolactone-co-D,L-lactide) within 35-63 g/mol, this study highlights that MCF7 cells can sense a 1000 times narrower relaxation time range (80-290 ms) compared to other studies by using a crosslinked hydrogel system. We propose that the rapid bulk relaxation response of the substrate promotes more reactive oxygen species generation in the formed semi-3D multicellular aggregates of breast cancer cells. Our finding sheds light on the potential role of bulk stress relaxation in a viscous-dominant viscoelastic matrix in controlling the cell cycle arrest depth of breast cancer cells.
Collapse
Affiliation(s)
- Mazaya Najmina
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba 305-0044, Japan
- Graduate School of Science and Engineering, University of Tsukuba, 1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Mitsuhiro Ebara
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba 305-0044, Japan
- Graduate School of Science and Engineering, University of Tsukuba, 1-1 Tennodai, Tsukuba 305-8577, Japan
- Graduate School of Industrial Science, Tokyo University of Science, 1-3 Kagurazaka, Shinjuku, Tokyo 162-8601, Japan
| | - Takahito Ohmura
- Research Center for Structural Materials, National Institute for Materials Science, 1-2-1 Sengen, Tsukuba 305-0047, Japan
| | - Koichiro Uto
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba 305-0044, Japan
- Correspondence:
| |
Collapse
|
10
|
Conti M, Bolzan I, Dal Zilio S, Parisse P, Andolfi L, Lazzarino M. Water-Air Interface to Mimic In Vitro Tumoral Cell Migration in Complex Micro-Environments. BIOSENSORS 2022; 12:822. [PMID: 36290959 PMCID: PMC9599853 DOI: 10.3390/bios12100822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
The long-known role of cell migration in physiological and pathological contexts still requires extensive research to be fully understood, mainly because of the intricate interaction between moving cells and their surroundings. While conventional assays fail to capture this complexity, recently developed 3D platforms better reproduce the cellular micro-environment, although often requiring expensive and time-consuming imaging approaches. To overcome these limitations, we developed a novel approach based on 2D micro-patterned substrates, compatible with conventional microscopy analysis and engineered to create micro-gaps with a length of 150 µm and a lateral size increasing from 2 to 8 µm, where a curved water-air interface is created on which cells can adhere, grow, and migrate. The resulting hydrophilic/hydrophobic interfaces, variable surface curvatures, spatial confinements, and size values mimic the complex micro-environment typical of the extracellular matrix in which aggressive cancer cells proliferate and migrate. The new approach was tested with two breast cancer cell lines with different invasive properties. We observed that invasive cells (MDA-MB-231) can align along the pattern and modify both their morphology and their migration rate according to the size of the water meniscus, while non-invasive cells (MCF-7) are only slightly respondent to the surrounding micro-environment. Moreover, the selected pattern highlighted a significative matrix deposition process connected to cell migration. Although requiring further optimizations, this approach represents a promising tool to investigate cell migration in complex environments.
Collapse
Affiliation(s)
- Martina Conti
- Department of Physics, University of Trieste, 34127 Trieste, Italy
- IOM-CNR, Institute of Materials Foundry—National Research Council, 34149 Trieste, Italy
| | - Ilaria Bolzan
- Department of Physics, University of Trieste, 34127 Trieste, Italy
- IOM-CNR, Institute of Materials Foundry—National Research Council, 34149 Trieste, Italy
| | - Simone Dal Zilio
- IOM-CNR, Institute of Materials Foundry—National Research Council, 34149 Trieste, Italy
| | - Pietro Parisse
- IOM-CNR, Institute of Materials Foundry—National Research Council, 34149 Trieste, Italy
| | - Laura Andolfi
- IOM-CNR, Institute of Materials Foundry—National Research Council, 34149 Trieste, Italy
| | - Marco Lazzarino
- IOM-CNR, Institute of Materials Foundry—National Research Council, 34149 Trieste, Italy
| |
Collapse
|
11
|
Chighizola M, Dini T, Marcotti S, D'Urso M, Piazzoni C, Borghi F, Previdi A, Ceriani L, Folliero C, Stramer B, Lenardi C, Milani P, Podestà A, Schulte C. The glycocalyx affects the mechanotransductive perception of the topographical microenvironment. J Nanobiotechnology 2022; 20:418. [PMID: 36123687 PMCID: PMC9484177 DOI: 10.1186/s12951-022-01585-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
The cell/microenvironment interface is the starting point of integrin-mediated mechanotransduction, but many details of mechanotransductive signal integration remain elusive due to the complexity of the involved (extra)cellular structures, such as the glycocalyx. We used nano-bio-interfaces reproducing the complex nanotopographical features of the extracellular matrix to analyse the glycocalyx impact on PC12 cell mechanosensing at the nanoscale (e.g., by force spectroscopy with functionalised probes). Our data demonstrates that the glycocalyx configuration affects spatio-temporal nanotopography-sensitive mechanotransductive events at the cell/microenvironment interface. Opposing effects of major glycocalyx removal were observed, when comparing flat and specific nanotopographical conditions. The excessive retrograde actin flow speed and force loading are strongly reduced on certain nanotopographies upon strong reduction of the native glycocalyx, while on the flat substrate we observe the opposite trend. Our results highlight the importance of the glycocalyx configuration in a molecular clutch force loading-dependent cellular mechanism for mechanosensing of microenvironmental nanotopographical features.
Collapse
Affiliation(s)
- Matteo Chighizola
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Tania Dini
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Stefania Marcotti
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Mirko D'Urso
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Claudio Piazzoni
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Francesca Borghi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Anita Previdi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Laura Ceriani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Claudia Folliero
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Brian Stramer
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Cristina Lenardi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Alessandro Podestà
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.
| | - Carsten Schulte
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.
| |
Collapse
|
12
|
Iazzolino G, Mendibil U, Arnaiz B, Ruiz-de-Angulo A, Azkargorta M, Uribe KB, Khatami N, Elortza F, Olalde B, Gomez-Vallejo V, Llop J, Abarrategi A. Decellularization of xenografted tumors provides cell-specific in vitro 3D environment. Front Oncol 2022; 12:956940. [PMID: 36059712 PMCID: PMC9434107 DOI: 10.3389/fonc.2022.956940] [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: 05/30/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
In vitro cell culture studies are common in the cancer research field, and reliable biomimetic 3D models are needed to ensure physiological relevance. In this manuscript, we hypothesized that decellularized xenograft tumors can serve as an optimal 3D substrate to generate a top-down approach for in vitro tumor modeling. Multiple tumor cell lines were xenografted and the formed solid tumors were recovered for their decellularization by several techniques and further characterization by histology and proteomics techniques. Selected decellularized tumor xenograft samples were seeded with the HCC1806 human triple-negative breast cancer (TNBC) basal-like subtype cell line, and cell behavior was compared among them and with other control 2D and 3D cell culture methods. A soft treatment using Freeze-EDTA-DNAse allows proper decellularization of xenografted tumor samples. Interestingly, proteomic data show that samples decellularized from TNBC basal-like subtype xenograft models had different extracellular matrix (ECM) compositions compared to the rest of the xenograft tumors tested. The in vitro recellularization of decellularized ECM (dECM) yields tumor-type–specific cell behavior in the TNBC context. Data show that dECM derived from xenograft tumors is a feasible substrate for reseeding purposes, thereby promoting tumor-type–specific cell behavior. These data serve as a proof-of-concept for further potential generation of patient-specific in vitro research models.
Collapse
Affiliation(s)
- Gaia Iazzolino
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Unai Mendibil
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
- TECNALIA, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Blanca Arnaiz
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Ane Ruiz-de-Angulo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Kepa B. Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Neda Khatami
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Beatriz Olalde
- TECNALIA, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Vanessa Gomez-Vallejo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Jordi Llop
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Ander Abarrategi
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- *Correspondence: Ander Abarrategi,
| |
Collapse
|
13
|
Mehta P, Rahman Z, Ten Dijke P, Boukany PE. Microfluidics meets 3D cancer cell migration. Trends Cancer 2022; 8:683-697. [PMID: 35568647 DOI: 10.1016/j.trecan.2022.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 01/12/2023]
Abstract
An early step of metastasis requires a complex and coordinated migration of invasive tumor cells into the surrounding tumor microenvironment (TME), which contains extracellular matrix (ECM). It is being appreciated that 3D matrix-based microfluidic models have an advantage over conventional in vitro and animal models to study tumor progression events. Recent microfluidic models have enabled recapitulation of key mechanobiological features present within the TME to investigate collective cancer cell migration and invasion. Microfluidics also allows for functional interrogation and therapeutic manipulation of specific steps to study the dynamic aspects of tumor progression. In this review, we focus on recent developments in cancer cell migration and how microfluidic strategies have evolved to address the physiological complexities of the TME to visualize migration modes adapted by various tumor cells.
Collapse
Affiliation(s)
- Pranav Mehta
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands; Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| |
Collapse
|
14
|
Yui Y, Kumai J, Watanabe K, Wakamatsu T, Sasagawa S. Lung fibrosis is a novel therapeutic target to suppress lung metastasis of osteosarcoma. Int J Cancer 2022; 151:739-751. [PMID: 35342929 DOI: 10.1002/ijc.34008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/20/2022] [Accepted: 03/22/2022] [Indexed: 11/07/2022]
Abstract
The prognosis of patients with metastatic and recurrent osteosarcoma has not improved over the last 30 years because no effective treatment strategy has been established for lung metastases. Although molecular-targeted drugs that modify the extracellular environment, such as anti-fibrotic agents, have been developed for cancer treatment, the suppressive effects of anti-fibrotic agents on osteosarcoma lung metastasis are unclear. Osteosarcomas need to adapt to considerable changes with respect to the stiffness of the environment and fibrosis during lung metastasis and may thus be vulnerable to fibrotic suppression as they originate at the site of a stiff bone with considerable fibrosis. In this study, we investigated whether fibrosis was a therapeutic target for suppressing osteosarcoma metastasis. Lung tissue samples from patients and a mouse model (LM8-Dunn model) showed that lung metastatic colonization of osteosarcoma cells proceeded with massive lung fibrosis. Metastatic osteosarcoma LM8 cells proliferated in a scaffold-dependent manner; the proliferation was less dependent on YAP-mediated mechanotransduction on soft polyacrylamide gels. The anti-fibrotic agents pirfenidone and nintedanib suppressed lung metastasis in the LM8-Dunn model. The osteosarcoma cells did not show increased proliferation, as reported in breast cancer, after continuous culture in a soft environment. We speculated that the anti-fibrotic agents were effective because the osteosarcoma cells remained scaffold-dependent in the soft tissue environment. Thus, anti-fibrotic strategies may be useful in suppressing lung metastasis of bone and soft tissue tumors with stiff primary sites such as those in osteosarcoma.
Collapse
Affiliation(s)
- Yoshihiro Yui
- Research Institute, Nozaki Tokushukai Hospital, Daito, Japan
| | - Jun Kumai
- Research Institute, Nozaki Tokushukai Hospital, Daito, Japan
| | - Kenta Watanabe
- Research Institute, Nozaki Tokushukai Hospital, Daito, Japan.,Department of Orthopedic Surgery, Toyama University Hospital, Toyama, Japan
| | - Toru Wakamatsu
- Department of Orthopedic Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Satoru Sasagawa
- Research Institute, Nozaki Tokushukai Hospital, Daito, Japan
| |
Collapse
|
15
|
Pamonag M, Hinson A, Burton EJ, Jafari N, Sales D, Babcock S, Basha R, Hu X, Kubow KE. Individual cells generate their own self-reinforcing contact guidance cues through local matrix fiber remodeling. PLoS One 2022; 17:e0265403. [PMID: 35333902 PMCID: PMC8956187 DOI: 10.1371/journal.pone.0265403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
Directed cell migration arises from cells following a microenvironmental gradient (e.g. of a chemokine) or polarizing feature (e.g. a linear structure). However cells not only follow, but in many cases, also generate directionality cues by modifying their microenvironment. This bi-directional relationship is seen in the alignment of extracellular matrix (ECM) fibers ahead of invading cell masses. The forces generated by many migrating cells cause fiber alignment, which in turn promotes further migration in the direction of fiber alignment via contact guidance and durotaxis. While this positive-feedback relationship has been widely described for cells invading en masse, single cells are also able to align ECM fibers, as well as respond to contact guidance and durotaxis cues, and should therefore exhibit the same relationship. In this study, we directly tested this hypothesis by studying the migration persistence of individual HT-1080 fibrosarcoma cells migrating in photocrosslinked collagen matrices with limited remodeling potential. Our results demonstrate that this positive-feedback relationship is indeed a fundamental aspect of cell migration in fibrillar environments. We observed that the cells’ inability to align and condense fibers resulted in a decrease in persistence relative to cells in native collagen matrices and even relative to isotropic (glass) substrates. Further experiments involving 2D collagen and electrospun polymer scaffolds suggest that substrates composed of rigid, randomly oriented fibers reduce cells’ ability to follow another directionality cue by forcing them to meander to follow the available adhesive area (i.e. fibers). Finally, our results demonstrate that the bi-directional relationship between cell remodeling and migration is not a “dimensionality” effect, but a fundamental effect of fibrous substrate structure.
Collapse
Affiliation(s)
- Michael Pamonag
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Abigail Hinson
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Elisha J. Burton
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Nojan Jafari
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Dominic Sales
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Sarah Babcock
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Rozlan Basha
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Xiaofeng Hu
- Department of Chemistry & Biochemistry and Center for Materials Science, James Madison University, Harrisonburg, Virginia, United States of America
| | - Kristopher E. Kubow
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
16
|
Microenvironmental stiffness mediates cytoskeleton re-organization in chondrocytes through laminin-FAK mechanotransduction. Int J Oral Sci 2022; 14:15. [PMID: 35277477 PMCID: PMC8917190 DOI: 10.1038/s41368-022-00165-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/07/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
AbstractMicroenvironmental biophysical factors play a fundamental role in controlling cell behaviors including cell morphology, proliferation, adhesion and differentiation, and even determining the cell fate. Cells are able to actively sense the surrounding mechanical microenvironment and change their cellular morphology to adapt to it. Although cell morphological changes have been considered to be the first and most important step in the interaction between cells and their mechanical microenvironment, their regulatory network is not completely clear. In the current study, we generated silicon-based elastomer polydimethylsiloxane (PDMS) substrates with stiff (15:1, PDMS elastomer vs. curing agent) and soft (45:1) stiffnesses, which showed the Young’s moduli of ~450 kPa and 46 kPa, respectively, and elucidated a new path in cytoskeleton re-organization in chondrocytes in response to changed substrate stiffnesses by characterizing the axis shift from the secreted extracellular protein laminin β1, focal adhesion complex protein FAK to microfilament bundling. We first showed the cellular cytoskeleton changes in chondrocytes by characterizing the cell spreading area and cellular synapses. We then found the changes of secreted extracellular linkage protein, laminin β1, and focal adhesion complex protein, FAK, in chondrocytes in response to different substrate stiffnesses. These two proteins were shown to be directly interacted by Co-IP and colocalization. We next showed that impact of FAK on the cytoskeleton organization by showing the changes of microfilament bundles and found the potential intermediate regulators. Taking together, this modulation axis of laminin β1-FAK-microfilament could enlarge our understanding about the interdependence among mechanosensing, mechanotransduction, and cytoskeleton re-organization.
Collapse
|
17
|
Kpeglo D, Hughes MD, Dougan L, Haddrick M, Knowles MA, Evans SD, Peyman SA. Modeling the mechanical stiffness of pancreatic ductal adenocarcinoma. Matrix Biol Plus 2022; 14:100109. [PMID: 35399702 PMCID: PMC8990173 DOI: 10.1016/j.mbplus.2022.100109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 01/18/2023] Open
Abstract
The PDAC stroma stiffness underlines its malignant behavior and drug resistance. 3D in vitro cultures must model the PDAC stroma to effectively drug efficacy. PSCs are responsible for the stroma, and its activity is increased with TGF-β. Develop a 3D culture model of PDAC, which includes PSCs and TGF-β. Assess the mechanical stiffness, stain for collagen, and investigate gemcitabine efficacy.
Despite improvements in the understanding of disease biology, pancreatic ductal adenocarcinoma (PDAC) remains the most malignant cancer of the pancreas. PDAC constitutes ∼95% of all pancreatic cancers, and it is highly resistant to therapeutics. The increased tissue rigidity, which stems from the rich fibrotic stroma in the tumor microenvironment, is central to disease development, physiology, and resistance to drug perfusion. Pancreatic stellate cells (PSCs) are responsible for overproduction of extracellular matrix in the fibrotic stroma, and this is exacerbated by the overexpression of transforming growth factor-β (TGF-β). However, there are few in vitro PDAC models, which include both PSCs and TGF-β or mimic in vivo-like tumor stiffness. In this study, we present a three-dimensional in vitro PDAC model, which includes PSCs and TGF-β, and recapitulates PDAC tissue mechanical stiffness. Using oscillatory shear rheology, we show the mechanical stiffness of the model is within range of the PDAC tissue stiffness by day 21 of culture and highlight that the matrix environment is essential to adequately capture PDAC disease. PDAC is a complex, aggressive disease with poor prognosis, and biophysically relevant in vitro PDAC models, which take into account tissue mechanics, will provide improved tumor models for effective therapeutic assessment.
Collapse
Affiliation(s)
- Delanyo Kpeglo
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
| | - Matthew D.G. Hughes
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK
| | - Lorna Dougan
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Block 35, Mereside Alderley Park, Alderley Edge, SK10 4TG, UK
| | - Margaret A. Knowles
- Leeds Institute of Medical Research at St James’s (LIMR), School of Medicine, University of Leeds, LS2 9 JT, UK
| | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Leeds Institute of Medical Research at St James’s (LIMR), School of Medicine, University of Leeds, LS2 9 JT, UK
- Corresponding author at: Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK.
| |
Collapse
|
18
|
Imparato G, Urciuolo F, Netti PA. Organ on Chip Technology to Model Cancer Growth and Metastasis. Bioengineering (Basel) 2022; 9:28. [PMID: 35049737 PMCID: PMC8772984 DOI: 10.3390/bioengineering9010028] [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: 11/24/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 12/18/2022] Open
Abstract
Organ on chip (OOC) has emerged as a major technological breakthrough and distinct model system revolutionizing biomedical research and drug discovery by recapitulating the crucial structural and functional complexity of human organs in vitro. OOC are rapidly emerging as powerful tools for oncology research. Indeed, Cancer on chip (COC) can ideally reproduce certain key aspects of the tumor microenvironment (TME), such as biochemical gradients and niche factors, dynamic cell-cell and cell-matrix interactions, and complex tissue structures composed of tumor and stromal cells. Here, we review the state of the art in COC models with a focus on the microphysiological systems that host multicellular 3D tissue engineering models and can help elucidate the complex biology of TME and cancer growth and progression. Finally, some examples of microengineered tumor models integrated with multi-organ microdevices to study disease progression in different tissues will be presented.
Collapse
Affiliation(s)
- Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; (F.U.); (P.A.N.)
| | - Francesco Urciuolo
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Department of Chemical, Materials and Industrial Production (DICMAPI), Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.leTecchio 80, 80125 Naples, Italy
| | - Paolo Antonio Netti
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Department of Chemical, Materials and Industrial Production (DICMAPI), Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.leTecchio 80, 80125 Naples, Italy
| |
Collapse
|
19
|
Huang M, Huang Y, LIU H, Tang Z, Chen Y, Huang Z, Xu S, Du J, Jia B. Hydrogels for Treatment of Oral and Maxillofacial Diseases: Current Research, Challenge, and Future Directions. Biomater Sci 2022; 10:6413-6446. [DOI: 10.1039/d2bm01036d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Oral and maxillofacial diseases such as infection and trauma often involve various organs and tissues, resulting in structural defects, dysfunctions and/or adverse effects on facial appearance. Hydrogels have been applied...
Collapse
|
20
|
Delinassios JG, Hoffman RM. The cancer-inhibitory effects of proliferating tumor-residing fibroblasts. Biochim Biophys Acta Rev Cancer 2021; 1877:188673. [PMID: 34953931 DOI: 10.1016/j.bbcan.2021.188673] [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: 10/12/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 11/19/2022]
Abstract
Initiation, local progression, and metastasis of cancer are associated with specific morphological, molecular, and functional changes in the extracellular matrix and the fibroblasts within the tumor microenvironment (TME). In the early stages of tumor development, fibroblasts are an obstacle that cancer cells must surpass or nullify to progress. Thus, in early tumor progression, specific signaling from cancer cells activates bio-pathways, which abolish the innate anticancer properties of fibroblasts and convert a high proportion of them to tumor-promoting cancer-associated fibroblasts (CAFs). Following this initial event, a wide spectrum of gene expression changes gradually leads to the development of a stromal fibroblast population with complex heterogeneity, creating fibroblast subtypes with characteristic profiles, which may alternate between being tumor-promotive and tumor-suppressive, topologically and chronologically in the TME. These fibroblast subtypes form the tumor's histological landscape including areas of cancer growth, inflammation, angiogenesis, invasion fronts, proliferating and non-proliferating fibroblasts, cancer-cell apoptosis, fibroblast apoptosis, and necrosis. These features reflect general deregulation of tissue homeostasis within the TME. This review discusses fundamental and current knowledge that has established the existence of anticancer fibroblasts within the various interacting elements of the TME. It is proposed that the maintenance of fibroblast proliferation is an essential parameter for the activation of their anticancer capacity, similar to that by which normal fibroblasts would be activated in wound repair, thus maintaining tissue homeostasis. Encouragement of research in this direction may render new means of cancer therapy and a greater understanding of tumor progression.
Collapse
Affiliation(s)
- John G Delinassios
- International Institute of Anticancer Research, 1(st) km Kapandritiou-Kalamou Rd., Kapandriti, 19014 Attica, Greece.
| | - Robert M Hoffman
- Department of Surgery, University of California, 9300 Campus Point Drive, La Jolla, CA 92037, USA; AntiCancer Inc., 7917 Ostrow St, San Diego, CA 92111, USA.
| |
Collapse
|
21
|
Zhang K, Feng Q, Fang Z, Gu L, Bian L. Structurally Dynamic Hydrogels for Biomedical Applications: Pursuing a Fine Balance between Macroscopic Stability and Microscopic Dynamics. Chem Rev 2021; 121:11149-11193. [PMID: 34189903 DOI: 10.1021/acs.chemrev.1c00071] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Owing to their unique chemical and physical properties, hydrogels are attracting increasing attention in both basic and translational biomedical studies. Although the classical hydrogels with static networks have been widely reported for decades, a growing number of recent studies have shown that structurally dynamic hydrogels can better mimic the dynamics and functions of natural extracellular matrix (ECM) in soft tissues. These synthetic materials with defined compositions can recapitulate key chemical and biophysical properties of living tissues, providing an important means to understanding the mechanisms by which cells sense and remodel their surrounding microenvironments. This review begins with the overall expectation and design principles of dynamic hydrogels. We then highlight recent progress in the fabrication strategies of dynamic hydrogels including both degradation-dependent and degradation-independent approaches, followed by their unique properties and use in biomedical applications such as regenerative medicine, drug delivery, and 3D culture. Finally, challenges and emerging trends in the development and application of dynamic hydrogels are discussed.
Collapse
Affiliation(s)
- Kunyu Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Qian Feng
- Bioengineering College, Chongqing University, Chongqing 400044, People's Republic of China
| | - Zhiwei Fang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, People's Republic of China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, People's Republic of China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, People's Republic of China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China
| |
Collapse
|
22
|
Neutrally charged self-assembling peptide hydrogel recapitulates in vitro mechanisms of breast cancer progression. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112200. [PMID: 34225853 DOI: 10.1016/j.msec.2021.112200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/05/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022]
Abstract
Self-assembling peptide hydrogels (SAPH) are a popular biomaterial due to their biocompatibility with a wide range of cell types, synthetic design, structural properties that provide a more accurate 3D microenvironment, and potential for cell- and/or drug-delivery system. Mimicking solid tumors in vitro using hydrogels is one method of testing anti-cancer drug efficacy and observing cancerous cell-ECM interactions within a 3D system. In this study, a SAPH, PeptiGel®Alpha1, was used to model in vitro the 3D breast tumor microenvironment. PeptiGel®Alpha1 is composed of entangled nanofibers with consistent diameter and mechanical properties similar to breast cancer that more accurately mimic the stiffness of breast tumor tissue than Matrigel® or collagen type I. PeptiGel®Alpha1 supported the viability and growth of the breast cancer cell lines MCF-7 and MDA-MB-231 and recapitulated key features of solid tumors such as hypoxia and invasion. MCF-7 cells in the hydrogels formed large spheroids resembling acini, while MDA-MB-231 remained dispersed. When treated with tamoxifen, PeptiGel®Alpha1 acted as a barrier, providing drug penetration geometry similar to that in vivo, providing better prediction of the drug effect. Finally, it was observed that MCF-7 cells engulfed the peptide matrix after 14 days, highlighting a potential use in drug delivery. PeptiGel®Alpha1 is a suitable platform for in vitro modeling of breast cancer.
Collapse
|
23
|
Fibroblast MMP14-Dependent Collagen Processing Is Necessary for Melanoma Growth. Cancers (Basel) 2021; 13:cancers13081984. [PMID: 33924099 PMCID: PMC8074311 DOI: 10.3390/cancers13081984] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Matrix metalloproteinases (MMPs) were considered as targets for the treatment of various cancers. However, initial trials using broad inhibitors to MMPs have failed, partly attributed to the contrasting functions of these proteases acting as tumor promoters and suppressors, among other reasons. Our data now suggest that specific inhibition of MMP14 might represent a more specific approach, as loss of this protease in fibroblasts resulted in reduced growth of grafted melanomas. Here, we found that deletion of MMP14 in fibroblasts generates a matrix-rich environment that reduces tumor vascularization and melanoma cell proliferation. In in vitro and ex vivo assays, we showed that the latter is mediated by stiffening of the tissue due to collagen accumulation. Additionally, in vivo, we show that independently of MMP14 deletion, a collagen-rich stiff matrix inhibits the growth of melanomas. Abstract Skin homeostasis results from balanced synthesis and degradation of the extracellular matrix in the dermis. Deletion of the proteolytic enzyme MMP14 in dermal fibroblasts (MMP14Sf−/−) leads to a fibrotic skin phenotype with the accumulation of collagen type I, resulting from impaired proteolysis. Here, we show that melanoma growth in these mouse fibrotic dermal samples was decreased, paralleled by reduced tumor cell proliferation and vessel density. Using atomic force microscopy, we found increased peritumoral matrix stiffness of early but not late melanomas in the absence of fibroblast-derived MMP14. However, total collagen levels were increased at late melanoma stages in MMP14Sf−/− mice compared to controls. In ex vivo invasion assays, melanoma cells formed smaller tumor islands in MMP14Sf−/− skin, indicating that MMP14-dependent matrix accumulation regulates tumor growth. In line with these data, in vitro melanoma cell growth was inhibited in high collagen 3D spheroids or stiff substrates. Most importantly, in vivo induction of fibrosis using bleomycin reduced melanoma tumor growth. In summary, we show that MMP14 expression in stromal fibroblasts regulates melanoma tumor progression by modifying the peritumoral matrix and point to collagen accumulation as a negative regulator of melanoma.
Collapse
|
24
|
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
|
25
|
Construction of cancer-on-a-chip for drug screening. Drug Discov Today 2021; 26:1875-1890. [PMID: 33731317 DOI: 10.1016/j.drudis.2021.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/16/2020] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Abstract
Cancer-on-a-chip has effectively contributed to the development of drug screening, holding great promise for more convenient and reliable drug development as well as personalized drug administration.
Collapse
|
26
|
The distribution of liver cancer stem cells correlates with the mechanical heterogeneity of liver cancer tissue. Histochem Cell Biol 2021; 156:47-58. [PMID: 33710418 DOI: 10.1007/s00418-021-01979-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 12/27/2022]
Abstract
The survival of cancer stem cells is usually limited to a specific tumor microenvironment, and this microenvironment plays a vital role in the development of tumors. The mechanical properties of the microenvironment differ in different regions of solid tumors. However, in solid tumors, whether the distribution of cancer stem cells relates to the mechanical microenvironment of different regions is still unclear. In this study, we undertook a biophysical and biochemical assessment of the changes in the mechanical properties of liver tissue during the progression of liver cancer and explored the distribution of liver cancer stem cells in liver cancer tissues. Our analysis confirmed previous observations that the stiffness of liver tissue gradually increased with the progress of fibrosis. In liver cancer tissues, we found obvious mechanical heterogeneity: the core of the tumor was soft, the invasive front tissue was the hardest, and the para-cancer tissue was in an intermediate state. Interestingly, the greatest number of liver cancer stem cells was found in the invasive front part of the tumor. We finally established that stroma stiffness correlated with the number of liver cancer stem cells. These findings indicate that the distribution of liver cancer stem cells correlates with the mechanical heterogeneity of liver cancer tissue. This result provides a theoretical basis for the development of targeted therapies against the mechanical microenvironment of liver cancer stem cells.
Collapse
|
27
|
Teixeira FC, Chaves S, Torres AL, Barrias CC, Bidarra SJ. Engineering a Vascularized 3D Hybrid System to Model Tumor-Stroma Interactions in Breast Cancer. Front Bioeng Biotechnol 2021; 9:647031. [PMID: 33791288 PMCID: PMC8006407 DOI: 10.3389/fbioe.2021.647031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/16/2021] [Indexed: 01/23/2023] Open
Abstract
The stromal microenvironment of breast tumors, namely the vasculature, has a key role in tumor development and metastatic spread. Tumor angiogenesis is a coordinated process, requiring the cooperation of cancer cells, stromal cells, such as fibroblasts and endothelial cells, secreted factors and the extracellular matrix (ECM). In vitro models capable of capturing such complex environment are still scarce, but are pivotal to improve success rates in drug development and screening. To address this challenge, we developed a hybrid alginate-based 3D system, combining hydrogel-embedded mammary epithelial cells (parenchymal compartment) with a porous scaffold co-seeded with fibroblasts and endothelial cells (vascularized stromal compartment). For the stromal compartment, we used porous alginate scaffolds produced by freeze-drying with particle leaching, a simple, low-cost and non-toxic approach that provided storable ready-to-use scaffolds fitting the wells of standard 96-well plates. Co-seeded endothelial cells and fibroblasts were able to adhere to the surface, spread and organize into tubular-like structures. For the parenchymal compartment, a designed alginate gel precursor solution load with mammary epithelial cells was added to the pores of pre-vascularized scaffolds, forming a hydrogel in situ by ionic crosslinking. The 3D hybrid system supports epithelial morphogenesis in organoids/tumoroids and endothelial tubulogenesis, allowing heterotypic cell-cell and cell-ECM interactions, while presenting excellent experimental tractability for whole-mount confocal microscopy, histology and mild cell recovery for down-stream analysis. It thus provides a unique 3D in vitro platform to dissect epithelial-stromal interactions and tumor angiogenesis, which may assist in the development of selective and more effective anticancer therapies.
Collapse
Affiliation(s)
- Filipa C Teixeira
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Sara Chaves
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ana Luísa Torres
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Cristina C Barrias
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sílvia J Bidarra
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
28
|
Phenotypic Plasticity of Cancer Cells Based on Remodeling of the Actin Cytoskeleton and Adhesive Structures. Int J Mol Sci 2021; 22:ijms22041821. [PMID: 33673054 PMCID: PMC7918886 DOI: 10.3390/ijms22041821] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023] Open
Abstract
There is ample evidence that, instead of a binary switch, epithelial-mesenchymal transition (EMT) in cancer results in a flexible array of phenotypes, each one uniquely suited to a stage in the invasion-metastasis cascade. The phenotypic plasticity of epithelium-derived cancer cells gives them an edge in surviving and thriving in alien environments. This review describes in detail the actin cytoskeleton and E-cadherin-based adherens junction rearrangements that cancer cells need to implement in order to achieve the advantageous epithelial/mesenchymal phenotype and plasticity of migratory phenotypes that can arise from partial EMT.
Collapse
|
29
|
Abstract
Caveolin-1 (CAV1) has long been implicated in cancer progression, and while widely accepted as an oncogenic protein, CAV1 also has tumor suppressor activity. CAV1 was first identified in an early study as the primary substrate of Src kinase, a potent oncoprotein, where its phosphorylation correlated with cellular transformation. Indeed, CAV1 phosphorylation on tyrosine-14 (Y14; pCAV1) has been associated with several cancer-associated processes such as focal adhesion dynamics, tumor cell migration and invasion, growth suppression, cancer cell metabolism, and mechanical and oxidative stress. Despite this, a clear understanding of the role of Y14-phosphorylated pCAV1 in cancer progression has not been thoroughly established. Here, we provide an overview of the role of Src-dependent phosphorylation of tumor cell CAV1 in cancer progression, focusing on pCAV1 in tumor cell migration, focal adhesion signaling and metabolism, and in the cancer cell response to stress pathways characteristic of the tumor microenvironment. We also discuss a model for Y14 phosphorylation regulation of CAV1 effector protein interactions via the caveolin scaffolding domain.
Collapse
|
30
|
Jo J, Abdi Nansa S, Kim DH. Molecular Regulators of Cellular Mechanoadaptation at Cell-Material Interfaces. Front Bioeng Biotechnol 2020; 8:608569. [PMID: 33364232 PMCID: PMC7753015 DOI: 10.3389/fbioe.2020.608569] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022] Open
Abstract
Diverse essential cellular behaviors are determined by extracellular physical cues that are detected by highly orchestrated subcellular interactions with the extracellular microenvironment. To maintain the reciprocity of cellular responses and mechanical properties of the extracellular matrix, cells utilize a variety of signaling pathways that transduce biophysical stimuli to biochemical reactions. Recent advances in the micromanipulation of individual cells have shown that cellular responses to distinct physical and chemical features of the material are fundamental determinants of cellular mechanosensation and mechanotransduction. In the process of outside-in signal transduction, transmembrane protein integrins facilitate the formation of focal adhesion protein clusters that are connected to the cytoskeletal architecture and anchor the cell to the substrate. The linkers of nucleoskeleton and cytoskeleton molecular complexes, collectively termed LINC, are critical signal transducers that relay biophysical signals between the extranuclear cytoplasmic region and intranuclear nucleoplasmic region. Mechanical signals that involve cytoskeletal remodeling ultimately propagate into the nuclear envelope comprising the nuclear lamina in assistance with various nuclear membrane proteins, where nuclear mechanics play a key role in the subsequent alteration of gene expression and epigenetic modification. These intracellular mechanical signaling cues adjust cellular behaviors directly associated with mechanohomeostasis. Diverse strategies to modulate cell-material interfaces, including alteration of surface rigidity, confinement of cell adhesive region, and changes in surface topology, have been proposed to identify cellular signal transduction at the cellular and subcellular levels. In this review, we will discuss how a diversity of alterations in the physical properties of materials induce distinct cellular responses such as adhesion, migration, proliferation, differentiation, and chromosomal organization. Furthermore, the pathological relevance of misregulated cellular mechanosensation and mechanotransduction in the progression of devastating human diseases, including cardiovascular diseases, cancer, and aging, will be extensively reviewed. Understanding cellular responses to various extracellular forces is expected to provide new insights into how cellular mechanoadaptation is modulated by manipulating the mechanics of extracellular matrix and the application of these materials in clinical aspects.
Collapse
Affiliation(s)
| | | | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| |
Collapse
|
31
|
Fopase R, Saxena V, Seal P, Borah J, Pandey LM. Yttrium iron garnet for hyperthermia applications: Synthesis, characterization and in-vitro analysis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111163. [DOI: 10.1016/j.msec.2020.111163] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 01/09/2023]
|
32
|
Shinsato Y, Doyle AD, Li W, Yamada KM. Direct comparison of five different 3D extracellular matrix model systems for characterization of cancer cell migration. Cancer Rep (Hoboken) 2020; 3:e1257. [PMID: 33085847 PMCID: PMC7941507 DOI: 10.1002/cnr2.1257] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) in vitro model systems can bridge the gap between regular two-dimensional cell culture and whole-animal studies. Analyses of cancer cell migration and invasion increasingly use differing 3D systems, which may produce conflicting findings. AIMS We directly compared different 3D extracellular matrix systems for studying cancer cell migration/invasion by analyzing cell morphologies and quantifying aspects of cell migration including speed and directional persistence using automated computer-based cell tracking. METHODS AND RESULTS We performed direct comparisons of five different 3D extracellular matrix cell culture systems using both HT1080 fibrosarcoma and MDA-MB-231 breast carcinoma cell lines. The reconstituted 3D systems included two types of collagen hydrogel and tissue matrix gel (TMG) vs cell-derived matrices extracted from cultured primary human or cancer-associated fibroblasts. The fibrillar matrix architecture of these systems differed. 3D rat tail collagen and TMG matrices had short, randomly oriented collagen fibrils; bovine collagen had long, larger fibril bundles; and the cell-derived matrices were strongly oriented. HT1080 cells displayed rounded morphologies in all three reconstituted 3D matrices but became spindle shaped in the two cell-derived matrices. MDA-MB-231 cell morphologies were elongated in all matrices. Quantitative measures of cell migration parameters differed markedly between the different types of 3D matrix. Comparing the reconstituted matrices, cells migrated the most rapidly and furthest in TMG. Comparing TMG with cell-derived matrices, cells migrated more efficiently in the cell-derived matrices. The most notable differences were in directional persistence of migration, which was greatest in the two cell-derived matrices. CONCLUSION The morphologies of matrix fibrils and cell shape, and particularly the efficiency and directionality of cell migration, differed substantially depending on the type of 3D matrix system. We suggest that it is important to employ the 3D model system that most closely resembles the matrix environment being studied for analyses of cancer cell migration and invasion.
Collapse
Affiliation(s)
- Yoshinari Shinsato
- Cell Biology Section, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMarylandUSA
| | - Andrew D. Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMarylandUSA
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneWashingtonUSA
| | - Kenneth M. Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial ResearchNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
33
|
Balion Z, Sipailaite E, Stasyte G, Vailionyte A, Mazetyte-Godiene A, Seskeviciute I, Bernotiene R, Phopase J, Jekabsone A. Investigation of Cancer Cell Migration and Proliferation on Synthetic Extracellular Matrix Peptide Hydrogels. Front Bioeng Biotechnol 2020; 8:773. [PMID: 33014989 PMCID: PMC7498748 DOI: 10.3389/fbioe.2020.00773] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/18/2020] [Indexed: 01/10/2023] Open
Abstract
Chemical and mechanical properties of a tumor microenvironment are essential players in cancer progression, and it is important to precisely control the extracellular conditions while designing cancer in vitro models. The study investigates synthetic hydrogel matrices from multi-arm polyethylene glycol (PEG) functionalized with collagen-like peptide (CLP) CG(PKG)4(POG)4(DOG)4 alone and conjugated with either cell adhesion peptide RGD (mimicking fibronectin) or IKVAV (mimicking laminin). Human glioblastoma HROG36, rat C6 glioma cells, and A375 human melanoma cells were grown on the hydrogels and monitored for migration, proliferation, projected cell area, cell shape index, size and number, distribution of focal contacts in individual cells, and focal adhesion number. PEG-CLP-RGD induced migration of both glioma cell lines and also stimulated proliferation (assessed as metabolic activity) of HROG36 cells. Migration of C6 cells were also stimulated by PEG-CLP-IKVAV. These responses strongly correlated with the changes in adhesion and morphology parameters of individual cells – projected cell area, cell shape index, and focal contact number. Melanoma A375 cell proliferation was increased by PEG-CLP-RGD, and this was accompanied by a decrease in cell shape index. However, neither RGD nor IKVAV conjugated to PEG-CLP stimulated migratory capacity of A375 cells. Taken together, the study presents synthetic scaffolds with extracellular matrix (ECM)-mimicking peptides that allow for the exploration of the effect of ECM signaling to cancer cells.
Collapse
Affiliation(s)
- Zbigniev Balion
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Emilija Sipailaite
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Gabija Stasyte
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Agne Vailionyte
- Ferentis UAB, Vilnius, Lithuania.,Department of Nanoengineering, Center for Physical Sciences and Technology, Vilnius, Lithuania
| | - Airina Mazetyte-Godiene
- Ferentis UAB, Vilnius, Lithuania.,Department of Nanoengineering, Center for Physical Sciences and Technology, Vilnius, Lithuania
| | - Ieva Seskeviciute
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa Bernotiene
- Laboratory of Molecular Neurobiology, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Jaywant Phopase
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Aiste Jekabsone
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Laboratory of Molecular Neurobiology, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
34
|
Najmina M, Uto K, Ebara M. Fluidic substrate as a tool to probe breast cancer cell adaptive behavior in response to fluidity level. Polym J 2020. [DOI: 10.1038/s41428-020-0345-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Gaetani R, Zizzi EA, Deriu MA, Morbiducci U, Pesce M, Messina E. When Stiffness Matters: Mechanosensing in Heart Development and Disease. Front Cell Dev Biol 2020; 8:334. [PMID: 32671058 PMCID: PMC7326078 DOI: 10.3389/fcell.2020.00334] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
During embryonic morphogenesis, the heart undergoes a complex series of cellular phenotypic maturations (e.g., transition of myocytes from proliferative to quiescent or maturation of the contractile apparatus), and this involves stiffening of the extracellular matrix (ECM) acting in concert with morphogenetic signals. The maladaptive remodeling of the myocardium, one of the processes involved in determination of heart failure, also involves mechanical cues, with a progressive stiffening of the tissue that produces cellular mechanical damage, inflammation, and ultimately myocardial fibrosis. The assessment of the biomechanical dependence of the molecular machinery (in myocardial and non-myocardial cells) is therefore essential to contextualize the maturation of the cardiac tissue at early stages and understand its pathologic evolution in aging. Because systems to perform multiscale modeling of cellular and tissue mechanics have been developed, it appears particularly novel to design integrated mechano-molecular models of heart development and disease to be tested in ex vivo reconstituted cells/tissue-mimicking conditions. In the present contribution, we will discuss the latest implication of mechanosensing in heart development and pathology, describe the most recent models of cell/tissue mechanics, and delineate novel strategies to target the consequences of heart failure with personalized approaches based on tissue engineering and induced pluripotent stem cell (iPSC) technologies.
Collapse
Affiliation(s)
- Roberto Gaetani
- Department of Molecular Medicine, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Rome, Italy.,Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, San Diego, CA, United States
| | - Eric Adriano Zizzi
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Marco Agostino Deriu
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Umberto Morbiducci
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Maurizio Pesce
- Tissue Engineering Research Unit, "Centro Cardiologico Monzino," IRCCS, Milan, Italy
| | - Elisa Messina
- Department of Maternal, Infantile, and Urological Sciences, "Umberto I" Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
36
|
Yang Z, Xu H, Zhao X. Designer Self-Assembling Peptide Hydrogels to Engineer 3D Cell Microenvironments for Cell Constructs Formation and Precise Oncology Remodeling in Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903718. [PMID: 32382486 PMCID: PMC7201262 DOI: 10.1002/advs.201903718] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/08/2020] [Indexed: 02/05/2023]
Abstract
Designer self-assembling peptides form the entangled nanofiber networks in hydrogels by ionic-complementary self-assembly. This type of hydrogel has realistic biological and physiochemical properties to serve as biomimetic extracellular matrix (ECM) for biomedical applications. The advantages and benefits are distinct from natural hydrogels and other synthetic or semisynthetic hydrogels. Designer peptides provide diverse alternatives of main building blocks to form various functional nanostructures. The entangled nanofiber networks permit essential compositional complexity and heterogeneity of engineering cell microenvironments in comparison with other hydrogels, which may reconstruct the tumor microenvironments (TMEs) in 3D cell cultures and tissue-specific modeling in vitro. Either ovarian cancer progression or recurrence and relapse are involved in the multifaceted TMEs in addition to mesothelial cells, fibroblasts, endothelial cells, pericytes, immune cells, adipocytes, and the ECM. Based on the progress in common hydrogel products, this work focuses on the diverse designer self-assembling peptide hydrogels for instructive cell constructs in tissue-specific modeling and the precise oncology remodeling for ovarian cancer, which are issued by several research aspects in a 3D context. The advantages and significance of designer peptide hydrogels are discussed, and some common approaches and coming challenges are also addressed in current complex tumor diseases.
Collapse
Affiliation(s)
- Zehong Yang
- West China School of Basic Medical Sciences and Forensic MedicineSichuan UniversityChengduSichuan610041P. R. China
- Institute for Nanobiomedical Technology and Membrane BiologyWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Hongyan Xu
- GL Biochem (Shanghai) Ltd.519 Ziyue Rd.Shanghai200241P. R. China
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane BiologyWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
- Wenzhou InstituteUniversity of Chinese Academy of Sciences (Wenzhou Institute of Biomaterials & Engineering)WenzhouZhejiang325001P. R. China
| |
Collapse
|
37
|
Kusiak AA, Szopa MD, Jakubowska MA, Ferdek PE. Signaling in the Physiology and Pathophysiology of Pancreatic Stellate Cells - a Brief Review of Recent Advances. Front Physiol 2020; 11:78. [PMID: 32116785 PMCID: PMC7033654 DOI: 10.3389/fphys.2020.00078] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
The interest in pancreatic stellate cells (PSCs) has been steadily growing over the past two decades due mainly to the central role these cells have in the desmoplastic reaction associated with diseases of the pancreas, such as pancreatitis or pancreatic cancer. In recent years, the scientific community has devoted substantial efforts to understanding the signaling pathways that govern PSC activation and interactions with neoplastic cells. This mini review aims to summarize some very recent findings on signaling in PSCs and highlight their impact to the field.
Collapse
Affiliation(s)
- Agnieszka A Kusiak
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mateusz D Szopa
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | | | - Pawel E Ferdek
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
38
|
Azadi S, Tafazzoli Shadpour M. The microenvironment and cytoskeletal remodeling in tumor cell invasion. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:257-289. [DOI: 10.1016/bs.ircmb.2020.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Papalazarou V, Zhang T, Paul NR, Juin A, Cantini M, Maddocks ODK, Salmeron-Sanchez M, Machesky LM. The creatine-phosphagen system is mechanoresponsive in pancreatic adenocarcinoma and fuels invasion and metastasis. Nat Metab 2020; 2:62-80. [PMID: 32694686 PMCID: PMC7617069 DOI: 10.1038/s42255-019-0159-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 12/11/2019] [Indexed: 01/01/2023]
Abstract
Pancreatic ductal adenocarcinoma is particularly metastatic, with dismal survival rates and few treatment options. Stiff fibrotic stroma is a hallmark of pancreatic tumours, but how stromal mechanosensing affects metastasis is still unclear. Here, we show that mechanical changes in the pancreatic cancer cell environment affect not only adhesion and migration, but also ATP/ADP and ATP/AMP ratios. Unbiased metabolomic analysis reveals that the creatine-phosphagen ATP-recycling system is a major mechanosensitive target. This system depends on arginine flux through the urea cycle, which is reflected by the increased incorporation of carbon and nitrogen from L-arginine into creatine and phosphocreatine on stiff matrix. We identify that CKB is a mechanosensitive transcriptional target of YAP, and thus it increases phosphocreatine production. We further demonstrate that the creatine-phosphagen system has a role in invasive migration, chemotaxis and liver metastasis of cancer cells.
Collapse
Affiliation(s)
- Vassilis Papalazarou
- University of Glasgow Centre for the Cellular Microenvironment, Glasgow, UK
- University of Glasgow Institute of Cancer Sciences, Glasgow, UK
- CRUK Beatson Institute, Glasgow, UK
| | - Tong Zhang
- University of Glasgow Institute of Cancer Sciences, Glasgow, UK
| | | | | | - Marco Cantini
- University of Glasgow Centre for the Cellular Microenvironment, Glasgow, UK
| | | | | | - Laura M Machesky
- University of Glasgow Institute of Cancer Sciences, Glasgow, UK.
- CRUK Beatson Institute, Glasgow, UK.
| |
Collapse
|
40
|
Cell-Based Mechanosensation, Epigenetics, and Non-Coding RNAs in Progression of Cardiac Fibrosis. Int J Mol Sci 2019; 21:ijms21010028. [PMID: 31861579 PMCID: PMC6982012 DOI: 10.3390/ijms21010028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/11/2019] [Accepted: 12/15/2019] [Indexed: 12/22/2022] Open
Abstract
The heart is par excellence the 'in-motion' organ in the human body. Compelling evidence shows that, besides generating forces to ensure continuous blood supply (e.g., myocardial contractility) or withstanding passive forces generated by flow (e.g., shear stress on endocardium, myocardial wall strain, and compression strain at the level of cardiac valves), cells resident in the heart respond to mechanical cues with the activation of mechanically dependent molecular pathways. Cardiac stromal cells, most commonly named cardiac fibroblasts, are central in the pathologic evolution of the cardiovascular system. In their normal function, these cells translate mechanical cues into signals that are necessary to renew the tissues, e.g., by continuously rebuilding the extracellular matrix being subjected to mechanical stress. In the presence of tissue insults (e.g., ischemia), inflammatory cues, or modifiable/unmodifiable risk conditions, these mechanical signals may be 'misinterpreted' by cardiac fibroblasts, giving rise to pathology programming. In fact, these cells are subject to changing their phenotype from that of matrix renewing to that of matrix scarring cells-the so-called myo-fibroblasts-involved in cardiac fibrosis. The links between alterations in the abilities of cardiac fibroblasts to 'sense' mechanical cues and molecular pathology programming are still under investigation. On the other hand, various evidence suggests that cell mechanics may control stromal cells phenotype by modifying the epigenetic landscape, and this involves specific non-coding RNAs. In the present contribution, we will provide examples in support of this more integrated vision of cardiac fibrotic progression based on the decryption of mechanical cues in the context of epigenetic and non-coding RNA biology.
Collapse
|
41
|
Mohan V, Das A, Sagi I. Emerging roles of ECM remodeling processes in cancer. Semin Cancer Biol 2019; 62:192-200. [PMID: 31518697 DOI: 10.1016/j.semcancer.2019.09.004] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/01/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Extracellular matrix (ECM) plays a central and dynamic role in the creation of tumor microenvironment. Herein we discuss the emerging biophysical and biochemical aspects of ECM buildup and proteolysis in cancer niche formation. Dysregulated ECM remodeling by cancer cells facilitate irreversible proteolysis and crosslinking, which in turn influence cell signaling, micro environmental cues, angiogenesis and tissue biomechanics. Further, we introduce the emerging roles of cancer microbiome in aberrant tumor ECM remodeling and membrane bound nano-sized vesicles called exosomes in creation of distant pre-metastatic niches. A detailed molecular and biophysical understanding of the ECM morphologies and its components such as key enzymes, structural and signaling molecules are critical in identifying the next generation of therapeutic and diagnostic targets in cancer.
Collapse
Affiliation(s)
- Vishnu Mohan
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alakesh Das
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|