1
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation during Wound Closure. ACS Biomater Sci Eng 2025; 11:2249-2262. [PMID: 40029610 DOI: 10.1021/acsbiomaterials.4c02019] [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] [Indexed: 03/05/2025]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. Extra domain A (EDA) Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, EDA Fn organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional coactivator, yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate whether YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity, but on stiffer substrates, they decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest that there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
2
|
Botticelli M, Metzcar J, Phillips T, Cox S, Keshavanarayana P, Spill F. A hybrid computational model of cancer spheroid growth with ribose-induced collagen stiffening. Front Bioeng Biotechnol 2025; 13:1515962. [PMID: 40271351 PMCID: PMC12014586 DOI: 10.3389/fbioe.2025.1515962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025] Open
Abstract
Metastasis, the leading cause of death in cancer patients, arises when cancer cells disseminate from a primary solid tumour to distant organs. Growth and invasion of the solid tumour often involve collective cell migration, which is profoundly influenced by cell-cell interactions and the extracellular matrix (ECM). The ECM's biochemical composition and mechanical properties, such as stiffness, regulate cancer cell behaviour and migration dynamics. Mathematical modelling serves as a pivotal tool for studying and predicting these complex dynamics, with hybrid discrete-continuous models offering a powerful approach by combining agent-based representations of cells with continuum descriptions of the surrounding microenvironment. In this study, we investigate the impact of ECM stiffness, modulated via ribose-induced collagen cross-linking, on cancer spheroid growth and invasion. We employed a hybrid discrete-continuous model implemented in PhysiCell to simulate spheroid dynamics, successfully replicating three-dimensional in vitro experiments. The model incorporates detailed representations of cell-cell and cell-ECM interactions, ECM remodelling, and cell proliferation. Our simulations align with experimental observations of two breast cancer cell lines, non-invasive MCF7 and invasive HCC 1954, under varying ECM stiffness conditions. The results demonstrate that increased ECM stiffness due to ribose-induced cross-linking inhibits spheroid invasion in invasive cells, whereas non-invasive cells remain largely unaffected. Furthermore, our simulations show that higher ECM degradation by the cells not only enables spheroid growth and invasion but also facilitates the formation of multicellular protrusions. Conversely, increasing the maximum speed that cells can reach due to cell-ECM interactions enhances spheroid growth while promoting single-cell invasion. This hybrid modelling approach enhances our understanding of the interplay between cancer cell migration, proliferation, and ECM mechanical properties, paving the way for future studies incorporating additional ECM characteristics and microenvironmental conditions.
Collapse
Affiliation(s)
- Margherita Botticelli
- School of Mathematics, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - John Metzcar
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
| | - Thomas Phillips
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| | - Susan Cox
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| | - Pradeep Keshavanarayana
- School of Mathematics, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Computational Medicine, University College London, London, United Kingdom
| | - Fabian Spill
- School of Mathematics, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Tavano S, Brückner DB, Tasciyan S, Tong X, Kardos R, Schauer A, Hauschild R, Heisenberg CP. BMP-dependent patterning of ectoderm tissue material properties modulates lateral mesendoderm cell migration during early zebrafish gastrulation. Cell Rep 2025; 44:115387. [PMID: 40057955 DOI: 10.1016/j.celrep.2025.115387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/24/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
Cell migration is a fundamental process during embryonic development. Most studies in vivo have focused on the migration of cells using the extracellular matrix (ECM) as their substrate for migration. In contrast, much less is known about how cells migrate on other cells, as found in early embryos when the ECM has not yet formed. Here, we show that lateral mesendoderm (LME) cells in the early zebrafish gastrula use the ectoderm as their substrate for migration. We show that the lateral ectoderm is permissive for the animal-pole-directed migration of LME cells, while the ectoderm at the animal pole halts it. These differences in permissiveness depend on the lateral ectoderm being more cohesive than the animal ectoderm, a property controlled by bone morphogenetic protein (BMP) signaling within the ectoderm. Collectively, these findings identify ectoderm tissue cohesion as one critical factor in regulating LME migration during zebrafish gastrulation.
Collapse
Affiliation(s)
- Stefania Tavano
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | - David B Brückner
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Saren Tasciyan
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Xin Tong
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Roland Kardos
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alexandra Schauer
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | |
Collapse
|
4
|
Clemens C, Gehring R, Riedl P, Pompe T. Matrix deformation and mechanotransduction as markers of breast cancer cell phenotype alteration at matrix interfaces. Biomater Sci 2025; 13:1578-1589. [PMID: 39960148 DOI: 10.1039/d4bm01589d] [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: 03/12/2025]
Abstract
The dissemination of metastatic cells from the primary tumor into the surrounding tissue is a key event in the progression of cancer. This process involves the migration of cells across defined tissue interfaces that separate the dense tumor tissue from the adjacent healthy tissue. Prior research showed that cell transmigration across collagen I matrix interfaces induces a switch towards a more aggressive phenotype including a change in directionality of migration and chemosensitivity correlated to increased DNA damage during transmigration. Hence, mechanical forces acting at the nucleus during transmigration are hypothesized to trigger phenotype switching. Here, we present results from a particle image velocimetry (PIV) based live cell analysis of breast cancer cell transmigration across sharp matrix interfaces constituted of two collagen type I networks with different pore sizes. We found strong and highly localized collagen network deformation caused by cellular forces at the moment of crossing interfaces from dense into open matrices. Additionally, an increased contractility of transmigrated cells was determined for cells with the switch phenotype. Moreover, studies on mechanotransductive signaling at the nucleus, emerin translocation and YAP activation, indicated a misregulation of these signals for transmigrated cells with altered phenotype. These findings show that matrix interfaces between networks of different pore sizes mechanically challenge invasive breast cancer cells during transmigration by a strong asymmetry of contracting forces, impeding nuclear mechanotransduction pathways, with a subsequent trigger of more aggressive phenotypes.
Collapse
Affiliation(s)
- Cornelia Clemens
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Rosa Gehring
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Philipp Riedl
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Tilo Pompe
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| |
Collapse
|
5
|
Yoon YM, Woo SM, Guim H, Kim JA. Effects of calciprotein particles on EMT induction in an in vitro 3D-cultured proximal tubule epithelial cell model of CKD. Biofactors 2025; 51:e70009. [PMID: 40055954 DOI: 10.1002/biof.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/18/2025] [Indexed: 05/13/2025]
Abstract
Calciprotein particles (CPPs) are blood-borne circulating nanoparticles composed of calcium phosphate and proteins that are known to exacerbate pathological processes such as chronic kidney disease-mineral bone disorder (CKD-MBD). Despite the significant interest in CKD-MBD pathogenesis, research directly addressing CPP-induced fibrosis in renal proximal tubules is rare, largely owing to the lack of suitable in vitro tissue models. Our study confirmed that 3D-cultured renal proximal tubule epithelial cells (PTECs) exhibited enhanced pathological characteristics compared to 2D-cultured PTECs when treated with CPPs, a key factor in CKD-MBD, and the uremic toxin. 3D-cultured PTECs under CKD-inducing conditions by CPPs were associated with epithelial-mesenchymal transition (EMT), mediated by transforming growth factor-β1 (TGF-β1), with notable changes in early EMT marker expression. Furthermore, this was attributed to increased expression of the calcium-sensing receptor (CASR), a receptor for CPPs, and activation of the downstream cell division control protein 42 (CDC42), leading to EMT progression. This study underscores the potential of PTEC-on-a-chip systems to serve as drug testing models, given the heightened sensitivity of these cells to external environments. This approach provides a better understanding of the pathological features of CKD and could contribute to the development of more effective in vitro models and therapeutics.
Collapse
MESH Headings
- Epithelial-Mesenchymal Transition/drug effects
- Kidney Tubules, Proximal/pathology
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/cytology
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/genetics
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/genetics
- Humans
- Calcium Phosphates/pharmacology
- Animals
- Receptors, Calcium-Sensing/genetics
- Receptors, Calcium-Sensing/metabolism
- Cells, Cultured
Collapse
Affiliation(s)
- Yeo Min Yoon
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Sang-Mi Woo
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Hwanuk Guim
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
- Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
6
|
Perucca A, Llonín AG, Benach OM, Hallopeau C, Rivas EI, Linares J, Garrido M, Sallent-Aragay A, Golde T, Colombelli J, Dalaka E, Linacero J, Cazorla M, Galan T, Pastor Viel J, Badenas X, Recort-Bascuas A, Comerma L, Fernandez-Nogueira P, Rovira A, Roca-Cusachs P, Albanell J, Trepat X, Calon A, Labernadie A. Micro Immune Response On-chip (MIRO) models the tumour-stroma interface for immunotherapy testing. Nat Commun 2025; 16:1279. [PMID: 39900918 PMCID: PMC11790944 DOI: 10.1038/s41467-025-56275-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/13/2025] [Indexed: 02/05/2025] Open
Abstract
Immunotherapies are beneficial for a considerable proportion of cancer patients, but ineffective in others. In vitro modelling of the complex interactions between cancer cells and their microenvironment could provide a path to understanding immune therapy sensitivity and resistance. Here we develop MIRO, a fully humanised in vitro platform to model the spatial organisation of the tumour/stroma interface and its interaction with immune cells. We find that stromal barriers are associated with immune exclusion and protect cancer cells from antibody-dependent cellular cytotoxicity, elicited by targeted therapy. We demonstrate that IL2-driven immunomodulation increases immune cell velocity and spreading to overcome stromal immunosuppression and restores anti-cancer response in refractory tumours. Collectively, our study underscores the translational value of MIRO as a powerful tool for exploring how the spatial organisation of the tumour microenvironment shapes the immune landscape and influences the responses to immunomodulating therapies.
Collapse
Affiliation(s)
- Alice Perucca
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Andrea Gómez Llonín
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius, Réduit, Mauritius
| | - Oriol Mañé Benach
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Clement Hallopeau
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Elisa I Rivas
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Jenniffer Linares
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Marta Garrido
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Anna Sallent-Aragay
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Tom Golde
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Julien Colombelli
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Eleni Dalaka
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Judith Linacero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Marina Cazorla
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Teresa Galan
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Jordi Pastor Viel
- Unitat de Tecnologia Mecànica, Centres Científics i Tecnològics, Universitat de Barcelona, Barcelona, Spain
| | - Xavier Badenas
- Unitat de Tecnologia Mecànica, Centres Científics i Tecnològics, Universitat de Barcelona, Barcelona, Spain
| | - Alba Recort-Bascuas
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Laura Comerma
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Pathology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain
| | | | - Ana Rovira
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.
- Institucio Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain.
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.
| | - Alexandre Calon
- Cancer Research Program, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain.
| | - Anna Labernadie
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigacion Principe Felipe, Valencia, Spain.
| |
Collapse
|
7
|
Chastney MR, Kaivola J, Leppänen VM, Ivaska J. The role and regulation of integrins in cell migration and invasion. Nat Rev Mol Cell Biol 2025; 26:147-167. [PMID: 39349749 DOI: 10.1038/s41580-024-00777-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 01/29/2025]
Abstract
Integrin receptors are the main molecular link between cells and the extracellular matrix (ECM) as well as mediating cell-cell interactions. Integrin-ECM binding triggers the formation of heterogeneous multi-protein assemblies termed integrin adhesion complexes (IACs) that enable integrins to transform extracellular cues into intracellular signals that affect many cellular processes, especially cell motility. Cell migration is essential for diverse physiological and pathological processes and is dysregulated in cancer to favour cell invasion and metastasis. Here, we discuss recent findings on the role of integrins in cell migration with a focus on cancer cell dissemination. We review how integrins regulate the spatial distribution and dynamics of different IACs, covering classical focal adhesions, emerging adhesion types and adhesion regulation. We discuss the diverse roles integrins have during cancer progression from cell migration across varied ECM landscapes to breaching barriers such as the basement membrane, and eventual colonization of distant organs.
Collapse
Affiliation(s)
- Megan R Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jasmin Kaivola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Veli-Matti Leppänen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Life Technologies, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Western Finnish Cancer Center (FICAN West), University of Turku, Turku, Finland.
- Foundation for the Finnish Cancer Institute, Helsinki, Finland.
| |
Collapse
|
8
|
Ramos-Rodriguez DH, Fok SW, Dorais CJ, Filler AC, Caserta M, Leach JK. Decellularized Extracellular Matrix Improves Mesenchymal Stromal Cell Spheroid Response to Chondrogenic Stimuli. Tissue Eng Part A 2025; 31:139-151. [PMID: 39556314 PMCID: PMC11971541 DOI: 10.1089/ten.tea.2024.0267] [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: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 11/19/2024] Open
Abstract
Cartilage regeneration is hindered due to the low proliferative capacity of chondrocytes and the avascular nature of cartilaginous tissue. Mesenchymal stromal cells (MSCs) are widely studied for cartilage tissue engineering, and the aggregation of MSCs into high-density cell spheroids facilitates chondrogenic differentiation due to increased cell-cell contact. Despite the promise of MSCs, the field would benefit from improved strategies to regulate the chondrogenic potential of MSCs differentiated from induced pluripotent stem cells (iPSCs), which are advantageous for their capacity to yield large numbers of required cells. We previously demonstrated the ability of MSC-secreted extracellular matrix (ECM) to promote MSC chondrogenic differentiation, but the combinatorial effect of iPSC-derived MSC (iMSC) spheroids, iMSC-derived decellularized ECM (idECM), and other stimuli (e.g., oxygen tension and transforming growth factor [TGF]-β) on chondrogenic potential has not been described. Similar to MSCs, iMSCs secreted a collagen-rich ECM. When incorporated into spheroids, idECM increased spheroid diameter and promoted chondrogenic differentiation. The combination of idECM loading, chondrogenic media, and hypoxia enhanced glycosaminoglycan (GAG) content 1.6-fold (40.9 ± 4.6 ng vs. 25.6 ± 3.3 ng, p < 0.05) in iMSC spheroids. Compared with active TGF-β1, the presentation of latent TGF-β1 resulted in greater GAG content (26.6 ± 1.8 ng vs. 41.9 ± 4.3 ng, p < 0.01). Finally, we demonstrated the capacity of individual spheroids to self-assemble into larger constructs and undergo both chondrogenic and hypertrophic differentiation when maintained in lineage-inducing media. These results highlight the potential of idECM to enhance the efficacy of chondrogenic stimuli for improved cartilage regeneration using human MSCs and iMSCs.
Collapse
Affiliation(s)
| | - Shierly W. Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Connor J. Dorais
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| | - Andrea C. Filler
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Mason Caserta
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| | - J. Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| |
Collapse
|
9
|
Mikhajlov O, Adar RM, Tătulea-Codrean M, Macé AS, Manzi J, Tabarin F, Battistella A, di Federico F, Joanny JF, Tran van Nhieu G, Bassereau P. Cell adhesion and spreading on fluid membranes through microtubules-dependent mechanotransduction. Nat Commun 2025; 16:1201. [PMID: 39885125 PMCID: PMC11782702 DOI: 10.1038/s41467-025-56343-6] [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: 12/10/2023] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Integrin clusters facilitate mechanical force transmission (mechanotransduction) and regulate biochemical signaling during cell adhesion. However, most studies have focused on rigid substrates. On fluid substrates like supported lipid bilayers (SLBs), integrin ligands are mobile, and adhesive complexes are traditionally thought unable to anchor for cell spreading. Here, we demonstrate that cells spread on SLBs coated with Invasin, a high-affinity integrin ligand. Unlike SLBs functionalized with RGD peptides, integrin clusters on Invasin-SLBs grow in size and complexity comparable to those on glass. While actomyosin contraction dominates adhesion maturation on stiff substrates, we find that on fluid SLBs, integrin mechanotransduction and cell spreading rely on dynein pulling forces along microtubules perpendicular to the membranes and microtubules pushing on adhesive complexes, respectively. These forces, potentially present on non-deformable surfaces, are revealed in fluid substrate systems. Supported by a theoretical model, our findings demonstrate a mechanical role for microtubules in integrin clustering.
Collapse
Affiliation(s)
- Oleg Mikhajlov
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France.
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198, Inserm U1280, 1 Avenue de la Terrasse, 91190, Gif-sur-Yvette, France.
- Laboratory of Biophysics and Cell Biology of Signaling, Biochemistry department, University of Geneva, 30 quai Ernest-Ansermet, 1211, Geneva, Switzerland.
| | - Ram M Adar
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
- Collège de France, 11 place Marcelin Berthelot, 75005, Paris, France
- Department of Physics, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Maria Tătulea-Codrean
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
- Collège de France, 11 place Marcelin Berthelot, 75005, Paris, France
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, CB3 0WA, UK
| | - Anne-Sophie Macé
- Institut Curie, Université PSL, CNRS UMR144, Paris, France
- Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, Université PSL, CNRS, Paris, France
| | - John Manzi
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
| | - Fanny Tabarin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
| | - Aude Battistella
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
| | - Fahima di Federico
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
| | - Jean-François Joanny
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
- Collège de France, 11 place Marcelin Berthelot, 75005, Paris, France
| | - Guy Tran van Nhieu
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198, Inserm U1280, 1 Avenue de la Terrasse, 91190, Gif-sur-Yvette, France
| | - Patricia Bassereau
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France.
| |
Collapse
|
10
|
Zhou W, Lin J, Wang Q, Wang X, Yao X, Yan Y, Sun W, Zhu Q, Zhang X, Wang X, Ji B, Ouyang H. Chromatin-site-specific accessibility: A microtopography-regulated door into the stem cell fate. Cell Rep 2025; 44:115106. [PMID: 39723890 DOI: 10.1016/j.celrep.2024.115106] [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: 04/17/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Biomaterials that mimic extracellular matrix topography are crucial in tissue engineering. Previous research indicates that certain biomimetic topography can guide stem cells toward multiple specific lineages. However, the mechanisms by which topographic cues direct stem cell differentiation remain unclear. Here, we demonstrate that microtopography influences nuclear tension in mesenchymal stem cells (MSCs), shaping chromatin accessibility and determining lineage commitment. On aligned substrates, MSCs exhibit high cytoskeletal tension along the fiber direction, creating anisotropic nuclear stress that opens chromatin sites for neurogenic, myogenic, and tenogenic genes via transcription factors like Nuclear receptor TLX (TLX). In contrast, random substrates induce isotropic nuclear stress, promoting chromatin accessibility for osteogenic and chondrogenic genes through Runt-related transcription factors (RUNX). Our findings reveal that aligned and random microtopographies direct site-specific chromatin stretch and lineage-specific gene expression, priming MSCs for distinct lineages. This study introduces a novel framework for understanding how topographic cues govern cell fate in tissue repair and regeneration.
Collapse
Affiliation(s)
- Wenyan Zhou
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; School of Medicine, Taizhou University, Taizhou, Zhejiang Province 318000, China
| | - Junxin Lin
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; School of Medicine, Taizhou University, Taizhou, Zhejiang Province 318000, China
| | - Qianchun Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325000, China
| | - Xianliu Wang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 200051, China
| | - Xudong Yao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province 322000, China
| | - Yiyang Yan
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China
| | - Wei Sun
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China
| | - Qiuwen Zhu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiaoan Zhang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiaozhao Wang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Baohua Ji
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, Zhejiang Province 310027, China; Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310027, China.
| | - Hongwei Ouyang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
11
|
Brunel LG, Long CM, Christakopoulos F, Cai B, Johansson PK, Singhal D, Enejder A, Myung D, Heilshorn SC. Interpenetrating networks of fibrillar and amorphous collagen promote cell spreading and hydrogel stability. Acta Biomater 2025; 193:128-142. [PMID: 39798635 PMCID: PMC11908676 DOI: 10.1016/j.actbio.2025.01.009] [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: 09/06/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Hydrogels composed of collagen, the most abundant protein in the human body, are widely used as scaffolds for tissue engineering due to their ability to support cellular activity. However, collagen hydrogels with encapsulated cells often experience bulk contraction due to cell-generated forces, and conventional strategies to mitigate this undesired deformation often compromise either the fibrillar microstructure or cytocompatibility of the collagen. To support the spreading of encapsulated cells while preserving the structural integrity of the gels, we present an interpenetrating network (IPN) of two distinct collagen networks with different crosslinking mechanisms and microstructures. First, a physically self-assembled collagen network preserves the fibrillar microstructure and enables the spreading of encapsulated human corneal mesenchymal stromal cells. Second, an amorphous collagen network covalently crosslinked with bioorthogonal chemistry fills the voids between fibrils and stabilizes the gel against cell-induced contraction. This collagen IPN balances the biofunctionality of natural collagen with the stability of covalently crosslinked, engineered polymers. Taken together, these data represent a new avenue for maintaining both the fiber-induced spreading of cells and the structural integrity of collagen hydrogels by leveraging an IPN of fibrillar and amorphous collagen networks. STATEMENT OF SIGNIFICANCE: Collagen hydrogels are widely used as scaffolds for tissue engineering due to their support of cellular activity. However, collagen hydrogels often undergo undesired changes in size and shape due to cell-generated forces, and conventional strategies to mitigate this deformation typically compromise either the fibrillar microstructure or cytocompatibility of the collagen. In this study, we introduce an innovative interpenetrating network (IPN) that combines physically self-assembled, fibrillar collagen-ideal for promoting cell adhesion and spreading-with covalently crosslinked, amorphous collagen-ideal for enhancing bulk hydrogel stability. Our IPN design maintains the native fibrillar structure of collagen while significantly improving resistance against cell-induced contraction, providing a promising solution to enhance the performance and reliability of collagen hydrogels for tissue engineering applications.
Collapse
Affiliation(s)
- Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Chris M Long
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Fotis Christakopoulos
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Betty Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Patrik K Johansson
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Diya Singhal
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - David Myung
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA; Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
12
|
Hernandez K, Nguyen CH, Rijal G. Asporin increases the extracellular matrix cross-links and inhibits the cancer cell migration. Tumour Biol 2025; 47:10104283241313441. [PMID: 40099523 DOI: 10.1177/10104283241313441] [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] [Indexed: 03/20/2025] Open
Abstract
BackgroundMigrating strategies of the triple-negative breast cancer (TNBC) together with its role in the establishment of tumor microenvironment (TME), supporting metastasis, have been extensively studied. Extracellular matrix (ECM) is a major player for the TME, establishing the 3D spatial networks with interconnected pores necessary for the mechano-physiological function of the cells. Certain collagen aligners and cross-linkers which are necessary for the formation and the stabilization of ECM networks, however, have not been studied either in normal or in abnormal tissues. Complexities in cell-cell and cell-matrix interactions, and different in types and ratios of ECM proteins in a TME challenge to reveal the precise function of a particular protein that is exhibited by special cells and if specifically present in insignificant amount. Cancer-associated fibroblasts (CAFs) predominantly occupy the major stroma of a solid tumor where they deposit extracellular proteins in the excessive amount compared to other tumor-associated cells. For example, the TNBC tumor itself is positive for asporin (ASPN) since CAFs are major ASPN exhibitors. However, the TNBC cells express it insignificantly.ObjectiveThe increase in ECM and its networks suppresses the metastasis.MethodsHere, we studied the expression of collagen type I and ASPN in CAFS and MDA-MB-231 (MM231), and evaluated the role of ASPN in collagen alignment and crosslinking.ResultsTNBC cells have an insignificant expression of ASPN and scanty collagen fibers, some of which aggregate to form the stiff deranged fibers, forming large-size pores in ECM of cancer-cell-dominant outer core of TNBC that support cancer cell invasion and metastasis. Exogenous ASPN and fibroblast-ASPN supported for the collagen alignment and crosslinking that established the small-size pores in the ECM, inhibiting the cancer cell invasion.ConclusionsThe collagen aligner and the cross-linker, ASPN increases the ECM networks and decreases the migration, and this preliminary study provides the hope that ASPN might be used as an anti-metastatic drug after its confirmation through extensive studies in animal, and positive outcomes through preclinical trials.
Collapse
Affiliation(s)
- Kimberly Hernandez
- Department of Medical Laboratory Sciences, Public Health and Nutrition Science, Tarleton State University, A Member of Texas A & M University System, Fort Worth, TX, USA
| | - Caitlin H Nguyen
- Department of Medical Laboratory Sciences, Public Health and Nutrition Science, Tarleton State University, A Member of Texas A & M University System, Fort Worth, TX, USA
| | - Girdhari Rijal
- Department of Medical Laboratory Sciences, Public Health and Nutrition Science, Tarleton State University, A Member of Texas A & M University System, Fort Worth, TX, USA
| |
Collapse
|
13
|
Zihni C. Phagocytosis by the retinal pigment epithelium: New insights into polarized cell mechanics. Bioessays 2025; 47:e2300197. [PMID: 39663766 DOI: 10.1002/bies.202300197] [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: 10/12/2023] [Accepted: 10/21/2024] [Indexed: 12/13/2024]
Abstract
The retinal pigment epithelium (RPE) is a specialized epithelium at the back of the eye that carries out a variety of functions essential for visual health. Recent studies have advanced our molecular understanding of one of the major functions of the RPE; phagocytosis of spent photoreceptor outer segments (POS). Notably, a mechanical link, formed between apical integrins bound to extracellular POS and the intracellular actomyosin cytoskeleton, is proposed to drive the internalization of POS. The process may involve a "nibbling" action, as an initial step, to sever outer segment tips. These insights have led us to hypothesize an "integrin adhesome-like" network, atypically assembled at apical membrane RPE-POS contacts. I propose that this hypothetical network orchestrates the complex membrane remodeling events required for particle internalization. Therefore, its analysis and characterization will likely lead to a more comprehensive understanding of the molecular mechanisms that control POS phagocytosis.
Collapse
Affiliation(s)
- Ceniz Zihni
- Faculty of Health & Life Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
14
|
Tignard P, Pottin K, Geeverding A, Doulazmi M, Cabrera M, Fouquet C, Liffran M, Fouchard J, Rosello M, Albadri S, Del Bene F, Trembleau A, Breau MA. Basement membranes are crucial for proper olfactory placode shape, position and boundary with the brain, and for olfactory axon development. eLife 2024; 12:RP92004. [PMID: 39713923 DOI: 10.7554/elife.92004] [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] [Indexed: 12/24/2024] Open
Abstract
Despite recent progress, the complex roles played by the extracellular matrix in development and disease are still far from being fully understood. Here, we took advantage of the zebrafish sly mutation which affects Laminin γ1, a major component of basement membranes, to explore its role in the development of the olfactory system. Following a detailed characterisation of Laminin distribution in the developing olfactory circuit, we analysed basement membrane integrity, olfactory placode and brain morphogenesis, and olfactory axon development in sly mutants, using a combination of immunochemistry, electron microscopy and quantitative live imaging of cell movements and axon behaviours. Our results point to an original and dual contribution of Laminin γ1-dependent basement membranes in organising the border between the olfactory placode and the adjacent brain: they maintain placode shape and position in the face of major brain morphogenetic movements, they establish a robust physical barrier between the two tissues while at the same time allowing the local entry of the sensory axons into the brain and their navigation towards the olfactory bulb. This work thus identifies key roles of Laminin γ1-dependent basement membranes in neuronal tissue morphogenesis and axon development in vivo.
Collapse
Affiliation(s)
- Pénélope Tignard
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8246), Inserm U1130, Institut de Biologie Paris-Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Karen Pottin
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Audrey Geeverding
- Imaging Facility, Institut de Biologie Paris-Seine (IBPS), Paris, France
| | - Mohamed Doulazmi
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8256), Institut de Biologie Paris-Seine (IBPS), Adaptation Biologique et Vieillissement, Paris, France
| | - Mélody Cabrera
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Coralie Fouquet
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Mathilde Liffran
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8246), Inserm U1130, Institut de Biologie Paris-Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Jonathan Fouchard
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
| | - Marion Rosello
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Shahad Albadri
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Filippo Del Bene
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Alain Trembleau
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR8246), Inserm U1130, Institut de Biologie Paris-Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Marie Anne Breau
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS UMR7622), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| |
Collapse
|
15
|
van der Net A, Rahman Z, Bordoloi AD, Muntz I, ten Dijke P, Boukany PE, Koenderink GH. EMT-related cell-matrix interactions are linked to states of cell unjamming in cancer spheroid invasion. iScience 2024; 27:111424. [PMID: 39717087 PMCID: PMC11665421 DOI: 10.1016/j.isci.2024.111424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/25/2024] [Accepted: 11/15/2024] [Indexed: 12/25/2024] Open
Abstract
Epithelial-to-mesenchymal transitions (EMT) and unjamming transitions provide two distinct pathways for cancer cells to become invasive, but it is still unclear to what extent these pathways are connected. Here, we addressed this question by performing 3D spheroid invasion assays on epithelial-like (A549) and mesenchymal-like (MV3) cancer cell lines in collagen-based hydrogels, where we varied both the invasive character of the cells and matrix porosity. We found that the onset time of invasion was correlated with the matrix porosity and vimentin levels, while the spheroid expansion rate correlated with MMP1 levels. Spheroids displayed solid-like (non-invasive) states in small-pore hydrogels and fluid-like (strand-based) or gas-like (disseminating cells) states in large-pore hydrogels or for mesenchymal-like cells. Our findings are consistent with different unjamming states as a function of cell motility and matrix confinement predicted in recent models for cancer invasion, but show that cell motility and matrix confinement are coupled via EMT-related matrix degradation.
Collapse
Affiliation(s)
- Anouk van der Net
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft 2629 HZ, the Netherlands
| | - Zaid Rahman
- Delft University of Technology, Department of Chemical Engineering, Delft 2629 HZ, the Netherlands
| | - Ankur D. Bordoloi
- Delft University of Technology, Department of Chemical Engineering, Delft 2629 HZ, the Netherlands
| | - Iain Muntz
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft 2629 HZ, the Netherlands
| | - Peter ten Dijke
- Leiden University Medical Center, Department of Cell and Chemical Biology and Oncode Institute, Leiden 2333 ZC, the Netherlands
| | - Pouyan E. Boukany
- Delft University of Technology, Department of Chemical Engineering, Delft 2629 HZ, the Netherlands
| | - Gijsje H. Koenderink
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft 2629 HZ, the Netherlands
| |
Collapse
|
16
|
Yang H, Yang J, Zheng X, Chen T, Zhang R, Chen R, Cao T, Zeng F, Liu Q. The Hippo Pathway in Breast Cancer: The Extracellular Matrix and Hypoxia. Int J Mol Sci 2024; 25:12868. [PMID: 39684583 DOI: 10.3390/ijms252312868] [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: 11/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
As one of the most prevalent malignant neoplasms among women globally, the optimization of therapeutic strategies for breast cancer has perpetually been a research hotspot. The tumor microenvironment (TME) is of paramount importance in the progression of breast cancer, among which the extracellular matrix (ECM) and hypoxia are two crucial factors. The alterations of these two factors are predominantly regulated by the Hippo signaling pathway, which promotes tumor invasiveness, metastasis, therapeutic resistance, and susceptibility. Hence, this review focuses on the Hippo pathway in breast cancer, specifically, how the ECM and hypoxia impact the biological traits and therapeutic responses of breast cancer. Moreover, the role of miRNAs in modulating ECM constituents was investigated, and hsa-miR-33b-3p was identified as a potential therapeutic target for breast cancer. The review provides theoretical foundations and potential therapeutic direction for clinical treatment strategies in breast cancer, with the aspiration of attaining more precise and effective treatment alternatives in the future.
Collapse
Affiliation(s)
- Hanyu Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Jiaxin Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xiang Zheng
- School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tianshun Chen
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tingting Cao
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
17
|
Chauvière A, Manifacier I, Verdier C, Chagnon G, Cheddadi I, Glade N, Stéphanou A. A biomechanical model for cell sensing and migration. Comput Methods Biomech Biomed Engin 2024:1-19. [PMID: 39535176 DOI: 10.1080/10255842.2024.2427112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/06/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
We developed an original computational model for cell deformation and migration capable of accounting for the cell sensitivity to the environment and its appropriate adaptation. This cell model is ultimately intended to be used to address tissue morphogenesis. Hence it has been designed to comply with four requirements: (1) the cell should be able to probe and sense its environment and respond accordingly; (2) the model should be easy to parametrize to adapt to different cell types; (3) the model should be able to extend to 3D cases; (4) simulations should be fast enough to integrate many interacting cells. The simulations carried out focused on two aspects: first, the general behaviour of the cell on a homogeneous substrate, as observed experimentally, for model validation. This enabled us to decipher the mechanisms by which the cell can migrate, highlighting respective influences of the adhesions lifetimes and their sensitivity to traction; second, it predicts the sensitivity of the cell to an anisotropic patterned substrate, in agreement with recently published experiments. The results show that mechanosensors simulated by the model make it possible to reproduce such experiments in terms of migration bias generated by the substrate anisotropy. Here again, the model provides a biomechanical explanation of this phenomenon, depending on cell-matrix interactions and adhesion maturation rate.
Collapse
Affiliation(s)
- Arnaud Chauvière
- VetAgro Sup, Grenoble INP, TIMC, Université Grenoble Alpes, CNRS, UMR 5525, Grenoble, France
| | - Ian Manifacier
- VetAgro Sup, Grenoble INP, TIMC, Université Grenoble Alpes, CNRS, UMR 5525, Grenoble, France
| | - Claude Verdier
- LIPhy, Université Grenoble Alpes, CNRS, Grenoble, France
| | - Grégory Chagnon
- VetAgro Sup, Grenoble INP, TIMC, Université Grenoble Alpes, CNRS, UMR 5525, Grenoble, France
| | - Ibrahim Cheddadi
- VetAgro Sup, Grenoble INP, TIMC, Université Grenoble Alpes, CNRS, UMR 5525, Grenoble, France
| | - Nicolas Glade
- VetAgro Sup, Grenoble INP, TIMC, Université Grenoble Alpes, CNRS, UMR 5525, Grenoble, France
| | - Angélique Stéphanou
- VetAgro Sup, Grenoble INP, TIMC, Université Grenoble Alpes, CNRS, UMR 5525, Grenoble, France
| |
Collapse
|
18
|
Huang Y, Zhou Z, Liu T, Tang S, Xin X. Exploring heterogeneous cell population dynamics in different microenvironments by novel analytical strategy based on images. NPJ Syst Biol Appl 2024; 10:129. [PMID: 39505883 PMCID: PMC11542073 DOI: 10.1038/s41540-024-00459-w] [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: 06/11/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Understanding the dynamic states and transitions of heterogeneous cell populations is crucial for addressing fundamental biological questions. High-content imaging provides rich datasets, but it remains increasingly difficult to integrate and annotate high-dimensional and time-resolved datasets to profile heterogeneous cell population dynamics in different microenvironments. Using hepatic stellate cells (HSCs) LX-2 as model, we proposed a novel analytical strategy for image-based integration and annotation to profile dynamics of heterogeneous cell populations in 2D/3D microenvironments. High-dimensional features were extracted from extensive image datasets, and cellular states were identified based on feature profiles. Time-series clustering revealed distinct temporal patterns of cell shape and actin cytoskeleton reorganization. We found LX-2 showed more complex membrane dynamics and contractile systems with an M-shaped actin compactness trend in 3D culture, while they displayed rapid spreading in early 2D culture. This image-based integration and annotation strategy enhances our understanding of HSCs heterogeneity and dynamics in complex extracellular microenvironments.
Collapse
Affiliation(s)
- Yihong Huang
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zidong Zhou
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Tianqi Liu
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shengnan Tang
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xuegang Xin
- Laboratory of Biophysics, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Wang R, Sui Y, Liu Q, Xiong Y, Li S, Guo W, Xu Y, Zhang S. Recent advances in extracellular matrix manipulation for kidney organoid research. Front Pharmacol 2024; 15:1472361. [PMID: 39568581 PMCID: PMC11576200 DOI: 10.3389/fphar.2024.1472361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024] Open
Abstract
The kidney plays a crucial role in maintaining the body's microenvironment homeostasis. However, current treatment options and therapeutic agents for chronic kidney disease (CKD) are limited. Fortunately, the advent of kidney organoids has introduced a novel in vitro model for studying kidney diseases and drug screening. Despite significant efforts has been leveraged to mimic the spatial-temporal dynamics of fetal renal development in various types of kidney organoids, there is still a discrepancy in cell types and maturity compared to native kidney tissue. The extracellular matrix (ECM) plays a crucial role in regulating cellular signaling, which ultimately affects cell fate decision. As a result, ECM can refine the microenvironment of organoids, promoting their efficient differentiation and maturation. This review examines the existing techniques for culturing kidney organoids, evaluates the strengths and weaknesses of various types of kidney organoids, and assesses the advancements and limitations associated with the utilization of the ECM in kidney organoid culture. Additionally, it presents a discussion on constructing specific physiological and pathological microenvironments using decellularized extracellular matrix during certain developmental stages or disease occurrences, aiding the development of kidney organoids and disease models.
Collapse
Affiliation(s)
- Ren Wang
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yufei Sui
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Qiuyan Liu
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yucui Xiong
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Shanshan Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wu Guo
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiwei Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Sheng Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Barone L, Cucchiara M, Palano MT, Bassani B, Gallazzi M, Rossi F, Raspanti M, Zecca PA, De Antoni G, Pagiatakis C, Papait R, Bernardini G, Bruno A, Gornati R. Dental pulp mesenchymal stem cell (DPSCs)-derived soluble factors, produced under hypoxic conditions, support angiogenesis via endothelial cell activation and generation of M2-like macrophages. J Biomed Sci 2024; 31:99. [PMID: 39491013 PMCID: PMC11533415 DOI: 10.1186/s12929-024-01087-6] [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: 04/12/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Cell therapy has emerged as a revolutionary tool to repair damaged tissues by restoration of an adequate vasculature. Dental Pulp stem cells (DPSC), due to their easy biological access, ex vivo properties, and ability to support angiogenesis have been largely explored in regenerative medicine. METHODS Here, we tested the capability of Dental Pulp Stem Cell-Conditioned medium (DPSC-CM), produced in normoxic (DPSC-CM Normox) or hypoxic (DPSC-CM Hypox) conditions, to support angiogenesis via their soluble factors. CMs were characterized by a secretome protein array, then used for in vivo and in vitro experiments. In in vivo experiments, DPSC-CMs were associated to an Ultimatrix sponge and injected in nude mice. After excision, Ultimatrix were assayed by immunohistochemistry, electron microscopy and flow cytometry, to evaluate the presence of endothelial, stromal, and immune cells. For in vitro procedures, DPSC-CMs were used on human umbilical-vein endothelial cells (HUVECs), to test their effects on cell adhesion, migration, tube formation, and on their capability to recruit human CD14+ monocytes. RESULTS We found that DPSC-CM Hypox exert stronger pro-angiogenic activities, compared with DPSC-CM Normox, by increasing the frequency of CD31+ endothelial cells, the number of vessels and hemoglobin content in the Ultimatrix sponges. We observed that Utimatrix sponges associated with DPSC-CM Hypox or DPSC-CM Normox shared similar capability to recruit CD45- stromal cells, CD45+ leukocytes, F4/80+ macrophages, CD80+ M1-macrophages and CD206+ M2-macropages. We also observed that DPSC-CM Hypox and DPSC-CM Normox have similar capabilities to support HUVEC adhesion, migration, induction of a pro-angiogenic gene signature and the generation of capillary-like structures, together with the ability to recruit human CD14+ monocytes. CONCLUSIONS Our results provide evidence that DPSCs-CM, produced under hypoxic conditions, can be proposed as a tool able to support angiogenesis via macrophage polarization, suggesting its use to overcome the issues and restrictions associated with the use of staminal cells.
Collapse
Affiliation(s)
- Ludovica Barone
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Martina Cucchiara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Matteo Gallazzi
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Federica Rossi
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Mario Raspanti
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| | - Piero Antonio Zecca
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| | - Gianluca De Antoni
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Christina Pagiatakis
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Roberto Papait
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Giovanni Bernardini
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Antonino Bruno
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy.
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy.
| | - Rosalba Gornati
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy.
| |
Collapse
|
21
|
Guo K, van den Beucken T. Advances in drug-induced liver injury research: in vitro models, mechanisms, omics and gene modulation techniques. Cell Biosci 2024; 14:134. [PMID: 39488681 PMCID: PMC11531151 DOI: 10.1186/s13578-024-01317-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Drug-induced liver injury (DILI) refers to drug-mediated damage to the structure and function of the liver, ranging from mild elevation of liver enzymes to severe hepatic insufficiency, and in some cases, progressing to liver failure. The mechanisms and clinical symptoms of DILI are diverse due to the varying combination of drugs, making clinical treatment and prevention complex. DILI has significant public health implications and is the primary reason for post-marketing drug withdrawals. The search for reliable preclinical models and validated biomarkers to predict and investigate DILI can contribute to a more comprehensive understanding of adverse effects and drug safety. In this review, we examine the progress of research on DILI, enumerate in vitro models with potential benefits, and highlight cellular molecular perturbations that may serve as biomarkers. Additionally, we discuss omics approaches frequently used to gather comprehensive datasets on molecular events in response to drug exposure. Finally, three commonly used gene modulation techniques are described, highlighting their application in identifying causal relationships in DILI. Altogether, this review provides a thorough overview of ongoing work and approaches in the field of DILI.
Collapse
Affiliation(s)
- Kaidi Guo
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands.
| | - Twan van den Beucken
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
22
|
Zambuto SG, Scott AK, Oyen ML. Beyond 2D: Novel biomaterial approaches for modeling the placenta. Placenta 2024; 157:55-66. [PMID: 38514278 PMCID: PMC11399328 DOI: 10.1016/j.placenta.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
This review considers fully three-dimensional biomaterial environments of varying complexity as these pertain to research on the placenta. The developments in placental cell sources are first considered, along with the corresponding maternal cells with which the trophoblast interact. We consider biomaterial sources, including hybrid and composite biomaterials. Properties and characterization of biomaterials are discussed in the context of material design for specific placental applications. The development of increasingly complicated three-dimensional structures includes examples of advanced fabrication methods such as microfluidic device fabrication and 3D bioprinting, as utilized in a placenta context. The review finishes with a discussion of the potential for in vitro, three-dimensional placenta research to address health disparities and sexual dimorphism, especially in light of the exciting recent changes in the regulatory environment for in vitro devices.
Collapse
Affiliation(s)
- Samantha G Zambuto
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Women's Health Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Adrienne K Scott
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Women's Health Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Michelle L Oyen
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Women's Health Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
23
|
Piazza F, Ravaglia B, Caporale A, Svetić A, Parisse P, Asaro F, Grassi G, Secco L, Sgarra R, Marsich E, Donati I, Sacco P. Elucidating the unexpected cell adhesive properties of agarose substrates. The effect of mechanics, fetal bovine serum and specific peptide sequences. Acta Biomater 2024; 189:286-297. [PMID: 39357634 DOI: 10.1016/j.actbio.2024.09.042] [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: 05/21/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
2D agarose substrates have recently been surprisingly shown to be permissive for cell adhesion, depending on their mechanics and the use of the adhesive proteins of fetal bovine serum (FBS) in the cell culture medium. Here, we elucidate how the cells exhibit two anchoring mechanisms depending on the amount of FBS. Under low FBS conditions, the cells recognize the surface-coupled adhesive sequences of fibronectin via the binding of the heterodimer α5β1 integrin. Functionality of the actomyosin axis and mechanoactivation of focal adhesion kinase (FAK) are essential for the stretching of the protein, thereby accessing the "synergy" PPSRN site and enhancing cell adhesion in combination with the downstream RGD motif. Under high FBS conditions, the specific peptide sequences are much less relevant as the adsorbed serum proteins conceal the coupled fibronectin and the cells recognize the adhesive protein vitronectin, which is constitutively present in FBS, via the binding of the heterodimer αvβ3 integrin. Similarly, the intracellular tension and FAK activity are decisive, which collectively indicate that the cells stretch the partially cryptic RGD site of vitronectin and thus make it more accessible for integrin binding. Both anchoring mechanisms only work properly if the agarose substrate is mechanically compliant in terms of linear stress-strain response, unraveling a critical balance between the mechanics of the agarose substrate and the presentation of the adhesive peptides. STATEMENT OF SIGNIFICANCE: In the context of biomaterial design, agarose hydrogels are known to lack intrinsic cell-adhesive peptide motifs and are therefore commonly used for the development of non-permissive 2D substrates. However, we unexpectedly found that agarose hydrogels can become permissive substrates for cell adhesion, depending on a compliant mechanical response of the substrate and the use of fetal bovine serum (FBS) as protein reservoir in the cell culture medium. We describe here two anchoring mechanisms that cells harness to adhere to agarose substrates, depending on the amount of FBS. Our results will have a major impact on the field of mechanobiology and shed light on the central role of FBS as a natural source of adhesive proteins that could promote cell anchoring.
Collapse
Affiliation(s)
- Francesco Piazza
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Beatrice Ravaglia
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Andrea Caporale
- CNR, Institute of Crystallography, I-34149 Trieste, Italy; CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", I-80134 Napoli, Italy
| | - Ana Svetić
- Department of Physics, University of Trieste, Via Alfonso Valerio 2, I-34127 Trieste, Italy; Elettra Sincrotrone Trieste S.C.p.A., s.s. 14km 1635 in Area Science Park, I-34149 Trieste, Italy
| | - Pietro Parisse
- Elettra Sincrotrone Trieste S.C.p.A., s.s. 14km 1635 in Area Science Park, I-34149 Trieste, Italy; Istituto Officina dei Materiali (CNR-IOM), Consiglio Nazionale delle Ricerche, s.s. 14km 1635 in Area Science Park, I-34149 Trieste, Italy
| | - Fioretta Asaro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, I-34127 Trieste, Italy
| | - Gabriele Grassi
- Department of Medical, Surgical and Health Science, University of Trieste, Strada di Fiume 447, I-34129 Trieste, Italy
| | - Luca Secco
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Eleonora Marsich
- Department of Medical, Surgical and Health Science, University of Trieste, Strada di Fiume 447, I-34129 Trieste, Italy
| | - Ivan Donati
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Pasquale Sacco
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy.
| |
Collapse
|
24
|
Rufin M, Nalbach M, Rakuš M, Fuchs M, Poik M, Schitter G, Thurner PJ, Andriotis OG. Methylglyoxal alters collagen fibril nanostiffness and surface potential. Acta Biomater 2024; 189:208-216. [PMID: 39218277 DOI: 10.1016/j.actbio.2024.08.039] [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: 05/07/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Collagen fibrils are fundamental to the mechanical strength and function of biological tissues. However, they are susceptible to changes from non-enzymatic glycation, resulting in the formation of advanced glycation end-products (AGEs) that are not reversible. AGEs accumulate with aging and disease and can adversely impact tissue mechanics and cell-ECM interactions. AGE-crosslinks have been related, on the one hand, to dysregulation of collagen fibril stiffness and damage and, on the other hand, to altered collagen net surface charge as well as impaired cell recognition sites. While prior studies using Kelvin probe force microscopy (KPFM) have shown the effect glycation has on collagen fibril surface potential (i.e., net charge), the combined effect on individual and isolated collagen fibril mechanics, hydration, and surface potential has not been documented. Here, we explore how methylglyoxal (MGO) treatment affects the mechanics and surface potential of individual and isolated collagen fibrils by utilizing atomic force microscopy (AFM) nanoindentation and KPFM. Our results reveal that MGO treatment significantly increases nanostiffness, alters surface potential, and modifies hydration characteristics at the collagen fibril level. These findings underscore the critical impact of AGEs on collagen fibril physicochemical properties, offering insights into pathophysiological mechanical and biochemical alterations with implications for cell mechanotransduction during aging and in diabetes. STATEMENT OF SIGNIFICANCE: Collagen fibrils are susceptible to glycation, the irreversible reaction of amino acids with sugars. Glycation affects the mechanical properties and surface chemistry of collagen fibrils with adverse alterations in biological tissue mechanics and cell-ECM interactions. Current research on glycation, at the level of individual collagen fibrils, is sparse and has focused either on collagen fibril mechanics, with contradicting evidence, or surface potential. Here, we utilized a multimodal approach combining Kelvin probe force (KPFM) and atomic force microscopy (AFM) to examine how methylglyoxal glycation induces structural, mechanical, and surface potential changes on the same individual and isolated collagen fibrils. This approach helps inform structure-function relationships at the level of individual collagen fibrils.
Collapse
Affiliation(s)
- Manuel Rufin
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Mathis Nalbach
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Maja Rakuš
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Magdalena Fuchs
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Mathias Poik
- Automation and Control Institute (ACIN), TU Wien, Gusshausstrasse 27-29, A-1040 Vienna, Austria
| | - Georg Schitter
- Automation and Control Institute (ACIN), TU Wien, Gusshausstrasse 27-29, A-1040 Vienna, Austria
| | - Philipp J Thurner
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria
| | - Orestis G Andriotis
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Gumpendorfer Strasse 7, A-1060 Vienna, Austria.
| |
Collapse
|
25
|
Rodríguez-Quesada L, Ramírez-Sánchez K, Formosa-Dague C, Dague E, Sáenz-Arce G, García-González CA, Vásquez-Sancho F, Avendaño-Soto E, Starbird-Pérez R. Evaluation of Conductive Porous Biobased Composites with Tunable Mechanical Properties for Potential Biological Applications. ACS OMEGA 2024; 9:43426-43437. [PMID: 39493987 PMCID: PMC11525745 DOI: 10.1021/acsomega.4c04391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024]
Abstract
In this work, starch-based porous cryogels with controlled mechanical and electrical properties were prepared for tissue engineering applications. The starch cryogels were formulated using κ-carrageenan, poly(vinyl alcohol) (PVA), and styrylpyridinium-substituted PVA (SbQ) into the composite. A conductive cryogel was polymerized by chemical oxidation of 3,4-ethylenedioxythiophene (EDOT) using iron(III) p-toluenesulfonate as a strategy to control the electrical properties. The physical, thermal, and mechanical properties were evaluated for the obtained composites. Macro- and nanoscale results confirmed the capability of tuning the mechanical properties of the material by the addition of biopolymers in different contents. The presence of κ-carrageenan significantly increased the storage modulus and decreased the damping effect in the formulations. The presence of PVA showed a plasticizing effect in the formulations, confirmed by the buffering effect and an increase in storage modulus. PVA-SBQ improved the mechanical properties by cross-linking. The addition of PEDOT increased the mechanical and electrical properties of the obtained materials.
Collapse
Affiliation(s)
- Laria Rodríguez-Quesada
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
- Master
Program in Medical Devices Engineering, Instituto Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
- Departamento
de Física, Facultad de Ciencias Exactas y Naturales, Universidad Nacional, Heredia 86-3000, Costa Rica
| | - Karla Ramírez-Sánchez
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
| | | | - Etienne Dague
- LAAS-CNRS,
CNRS, Université de Toulouse, 31400Toulouse, France
| | - Giovanni Sáenz-Arce
- Departamento
de Física, Facultad de Ciencias Exactas y Naturales, Universidad Nacional, Heredia 86-3000, Costa Rica
- Centro de
Investigación en Óptica y Nanofísica, Departamento
de Física, Universidad de Murcia, 30100 Murcia, Spain
| | - Carlos A. García-González
- Departamento
de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago
de Compostela, Spain
| | - Fabián Vásquez-Sancho
- Centro
de Investigación en Ciencia e Ingeniería de Materiales
(CICIMA), Universidad de Costa Rica, San José 11501-2060, Costa Rica
- School
of Physics, Universidad de Costa Rica, San José 11501-2060, Costa Rica
| | - Esteban Avendaño-Soto
- Centro
de Investigación en Ciencia e Ingeniería de Materiales
(CICIMA), Universidad de Costa Rica, San José 11501-2060, Costa Rica
- School
of Physics, Universidad de Costa Rica, San José 11501-2060, Costa Rica
| | - Ricardo Starbird-Pérez
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
| |
Collapse
|
26
|
Mistriotis P, Wisniewski EO, Si BR, Kalab P, Konstantopoulos K. Coordinated in confined migration: crosstalk between the nucleus and ion channel-mediated mechanosensation. Trends Cell Biol 2024; 34:809-825. [PMID: 38290913 PMCID: PMC11284253 DOI: 10.1016/j.tcb.2024.01.001] [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: 09/03/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Cell surface and intracellular mechanosensors enable cells to perceive different geometric, topographical, and physical cues. Mechanosensitive ion channels (MICs) localized at the cell surface and on the nuclear envelope (NE) are among the first to sense and transduce these signals. Beyond compartmentalizing the genome of the cell and its transcription, the nucleus also serves as a mechanical gauge of different physical and topographical features of the tissue microenvironment. In this review, we delve into the intricate mechanisms by which the nucleus and different ion channels regulate cell migration in confinement. We review evidence suggesting an interplay between macromolecular nuclear-cytoplasmic transport (NCT) and ionic transport across the cell membrane during confined migration. We also discuss the roles of the nucleus and ion channel-mediated mechanosensation, whether acting independently or in tandem, in orchestrating migratory mechanoresponses. Understanding nuclear and ion channel sensing, and their crosstalk, is critical to advancing our knowledge of cell migration in health and disease.
Collapse
Affiliation(s)
| | - Emily O Wisniewski
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bishwa R Si
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Pallotta I, Stec MJ, Schriver B, Golann DR, Considine K, Su Q, Barahona V, Napolitano JE, Stanley S, Garcia M, Feric NT, Durney KM, Aschar‐Sobbi R, Bays N, Shavlakadze T, Graziano MP. Electrical stimulation of biofidelic engineered muscle enhances myotube size, force, fatigue resistance, and induces a fast-to-slow-phenotype shift. Physiol Rep 2024; 12:e70051. [PMID: 39384537 PMCID: PMC11464147 DOI: 10.14814/phy2.70051] [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: 08/17/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 10/11/2024] Open
Abstract
Therapeutic development for skeletal muscle diseases is challenged by a lack of ex vivo models that recapitulate human muscle physiology. Here, we engineered 3D human skeletal muscle tissue in the Biowire II platform that could be maintained and electrically stimulated long-term. Increasing differentiation time enhanced myotube formation, modulated myogenic gene expression, and increased twitch and tetanic forces. When we mimicked exercise training by applying chronic electrical stimulation, the "exercised" skeletal muscle tissues showed increased myotube size and a contractility profile, fatigue resistance, and gene expression changes comparable to in vivo models of exercise training. Additionally, tissues also responded with expected physiological changes to known pharmacological treatment. To our knowledge, this is the first evidence of a human engineered 3D skeletal muscle tissue that recapitulates in vivo models of exercise. By recapitulating key features of human skeletal muscle, we demonstrated that the Biowire II platform may be used by the pharmaceutical industry as a model for identifying and optimizing therapeutic drug candidates that modulate skeletal muscle function.
Collapse
Affiliation(s)
| | | | | | | | | | - Qi Su
- Regeneron PharmaceuticalsTarrytownNew YorkUSA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation During Wound Closure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614581. [PMID: 39386582 PMCID: PMC11463502 DOI: 10.1101/2024.09.23.614581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. EDA Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, Fn EDA organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional co-activator, Yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure, but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane (PDMS) substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate if YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity but on stiffer substrates, decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Pennsylvania
| | | | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Pennsylvania
| | - Michael Kegel
- Department of Bioengineering, Temple University, Pennsylvania
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Pennsylvania
| | - Karin Wang
- Department of Bioengineering, Temple University, Pennsylvania
| |
Collapse
|
29
|
Brunel LG, Long CM, Christakopoulos F, Cai B, Johansson PK, Singhal D, Enejder A, Myung D, Heilshorn SC. Interpenetrating networks of fibrillar and amorphous collagen promote cell spreading and hydrogel stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612534. [PMID: 39345483 PMCID: PMC11429934 DOI: 10.1101/2024.09.11.612534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Hydrogels composed of collagen, the most abundant protein in the human body, are widely used as scaffolds for tissue engineering due to their ability to support cellular activity. However, collagen hydrogels with encapsulated cells often experience bulk contraction due to cell-generated forces, and conventional strategies to mitigate this undesired deformation often compromise either the fibrillar microstructure or cytocompatibility of the collagen. To support the spreading of encapsulated cells while preserving the structural integrity of the gels, we present an interpenetrating network (IPN) of two distinct collagen networks with different crosslinking mechanisms and microstructures. First, a physically self-assembled collagen network preserves the fibrillar microstructure and enables the spreading of encapsulated human corneal mesenchymal stromal cells. Second, an amorphous collagen network covalently crosslinked with bioorthogonal chemistry fills the voids between fibrils and stabilizes the gel against cell-induced contraction. This collagen IPN balances the biofunctionality of natural collagen with the stability of covalently crosslinked, engineered polymers. Taken together, these data represent a new avenue for maintaining both the fiber-induced spreading of cells and the structural integrity of collagen hydrogels by leveraging an IPN of fibrillar and amorphous collagen networks. Statement of significance Collagen hydrogels are widely used as scaffolds for tissue engineering due to their support of cellular activity. However, collagen hydrogels often undergo undesired changes in size and shape due to cell-generated forces, and conventional strategies to mitigate this deformation typically compromise either the fibrillar microstructure or cytocompatibility of the collagen. In this study, we introduce an innovative interpenetrating network (IPN) that combines physically self-assembled, fibrillar collagen-ideal for promoting cell adhesion and spreading-with covalently crosslinked, amorphous collagen-ideal for enhancing bulk hydrogel stability. Our IPN design maintains the native fibrillar structure of collagen while significantly improving resistance against cell-induced contraction, providing a promising solution to enhance the performance and reliability of collagen hydrogels for tissue engineering applications. Graphical abstract
Collapse
|
30
|
Duong VT, Nguyen HD, Luong NH, Chang CY, Lin CC. Photo-responsive decellularized small intestine submucosa hydrogels. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2401952. [PMID: 39525288 PMCID: PMC11546089 DOI: 10.1002/adfm.202401952] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Indexed: 11/16/2024]
Abstract
Decellularized small intestine submucosa (dSIS) is a promising biomaterial for promoting tissue regeneration. Isolated from the submucosal layer of animal jejunum, SIS is rich in extracellular matrix (ECM) proteins, including collagen, laminin, and fibronectin. Following mild decellularization, dSIS becomes an acellular matrix that supports cell adhesion, proliferation, and differentiation. Conventional dSIS matrix is usually obtained by thermal crosslinking, which yields a soft scaffold with low stability. To address these challenges, dSIS has been modified with methacrylate groups for photocrosslinking into stable hydrogels. However, dSIS has not been modified with clickable handles for orthogonal crosslinking. Here, we report the development of norbornene-modified dSIS, named dSIS-NB, via reacting amine groups of dSIS with carbic anhydride in acidic aqueous reaction conditions. Using triethylamine (TEA) as a mild base catalyst, we obtained high degrees of NB substitution on dSIS. In addition to describing the synthesis of dSIS-NB, we explored its adaptability in orthogonal hydrogel crosslinking and used dSIS-NB hydrogels for cancer and vascular tissue engineering. Impressively, compared with physically crosslinked dSIS and collagen matrices, orthogonally crosslinked dSIS-NB hydrogels supported rapid dissemination of cancer cells and superior vasculogenic and angiogenic properties. dSIS-NB was also exploited as a versatile bioink for 3D bioprinting applications.
Collapse
Affiliation(s)
- Van Thuy Duong
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Han Dang Nguyen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Ngoc Ha Luong
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Chun-Yi Chang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Paukner D, Jennings IR, Cyron CJ, Humphrey JD. Dynamic biaxial loading of vascular smooth muscle cell seeded tissue equivalents. J Mech Behav Biomed Mater 2024; 157:106639. [PMID: 38970943 DOI: 10.1016/j.jmbbm.2024.106639] [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: 03/26/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024]
Abstract
An intricate reciprocal relationship exists between adherent synthetic cells and their extracellular matrix (ECM). These cells deposit, organize, and degrade the ECM, which in turn influences cell phenotype via responses that include sensitivity to changes in the mechanical state that arises from changes in external loading. Collagen-based tissue equivalents are commonly used as simple but revealing model systems to study cell-matrix interactions. Nevertheless, few quantitative studies report changes in the forces that the cells establish and maintain in such gels under dynamic loading. Moreover, most prior studies have been limited to uniaxial experiments despite many soft tissues, including arteries, experiencing multiaxial loading in vivo. To begin to close this gap, we use a custom biaxial bioreactor to subject collagen gels seeded with primary aortic smooth muscle cells to different biaxial loading conditions. These conditions include cyclic loading with different amplitudes as well as different mechanical constraints at the boundaries of a cruciform sample. Irrespective of loading amplitude and boundary condition, similar mean steady-state biaxial forces emerged across all tests. Additionally, stiffness-force relationships assessed via intermittent equibiaxial force-extension tests showed remarkable similarity for ranges of forces to which the cells adapted during periods of cyclic loading. Taken together, these findings are consistent with a load-mediated homeostatic response by vascular smooth muscle cells.
Collapse
Affiliation(s)
- Daniel Paukner
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany; Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Geesthacht, Germany; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| | | | - Christian J Cyron
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany; Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Geesthacht, Germany
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
32
|
Blázquez-Carmona P, Ruiz-Mateos R, Barrasa-Fano J, Shapeti A, Martín-Alfonso JE, Domínguez J, Van Oosterwyck H, Reina-Romo E, Sanz-Herrera JA. Quantitative atlas of collagen hydrogels reveals mesenchymal cancer cell traction adaptation to the matrix nanoarchitecture. Acta Biomater 2024; 185:281-295. [PMID: 38992411 DOI: 10.1016/j.actbio.2024.07.002] [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: 02/26/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
Collagen-based hydrogels are commonly used in mechanobiology to mimic the extracellular matrix. A quantitative analysis of the influence of collagen concentration and properties on the structure and mechanics of the hydrogels is essential for tailored design adjustments for specific in vitro conditions. We combined focused ion beam scanning electron microscopy and rheology to provide a detailed quantitative atlas of the mechanical and nanoscale three-dimensional structural alterations that occur when manipulating different hydrogel's physicochemistry. Moreover, we study the effects of such alterations on the phenotype of breast cancer cells and their mechanical interactions with the extracellular matrix. Regardless of the microenvironment's pore size, porosity or mechanical properties, cancer cells are able to reach a stable mesenchymal-like morphology. Additionally, employing 3D traction force microscopy, a positive correlation between cellular tractions and ECM mechanics is observed up to a critical threshold, beyond which tractions plateau. This suggests that cancer cells in a stable mesenchymal state calibrate their mechanical interactions with the ECM to keep their migration and invasiveness capacities unaltered. STATEMENT OF SIGNIFICANCE: The paper presents a thorough study on the mechanical microenvironment in breast cancer cells during their interaction with collagen based hydrogels of different compositions. The hydrogels' microstructure were obtained using state-of-the-art 3D microscopy, namely focused ion beam-scanning electron microscope (FIB-SEM). FIB-SEM was originally applied in this work to reconstruct complex fibered collagen microstructures within the nanometer range, to obtain key microarchitectural parameters. The mechanical microenvironment of cells was recovered using Traction Force Microscopy (TFM). The obtained results suggest that cells calibrate tractions such that they depend on mechanical, microstructural and physicochemical characteristics of the hydrogels, hence revealing a steric hindrance. We hypothesize that cancer cells studied in this paper tune their mechanical state to keep their migration and invasiveness capacities unaltered.
Collapse
Affiliation(s)
- Pablo Blázquez-Carmona
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Raquel Ruiz-Mateos
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Jorge Barrasa-Fano
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Apeksha Shapeti
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - José Enrique Martín-Alfonso
- Escuela Técnica Superior de Ingeniería, Universidad de Huelva. Avda. de las Fuerzas Armadas s/n, 21007 Huelva, Spain
| | - Jaime Domínguez
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Hans Van Oosterwyck
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Esther Reina-Romo
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - José Antonio Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain.
| |
Collapse
|
33
|
Zhu T, Hu Y, Cui H, Cui H. 3D Multispheroid Assembly Strategies towards Tissue Engineering and Disease Modeling. Adv Healthc Mater 2024; 13:e2400957. [PMID: 38924326 DOI: 10.1002/adhm.202400957] [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: 03/13/2024] [Revised: 06/21/2024] [Indexed: 06/28/2024]
Abstract
Cell spheroids (esp. organoids) as 3D culture platforms are popular models for representing cell-cell and cell-extracellular matrix (ECM) interactions, bridging the gap between 2D cell cultures and natural tissues. 3D cell models with spatially organized multiple cell types are preferred for gaining comprehensive insights into tissue pathophysiology and constructing in vitro tissues and disease models because of the complexities of natural tissues. In recent years, an assembly strategy using cell spheroids (or organoids) as living building blocks has been developed to construct complex 3D tissue models with spatial organization. Here, a comprehensive overview of recent advances in multispheroid assembly studies is provided. The different mechanisms of the multispheroid assembly techniques, i.e., automated directed assembly, noncontact remote assembly, and programmed self-assembly, are introduced. The processing steps, advantages, and technical limitations of the existing methodologies are summarized. Applications of the multispheroid assembly strategies in disease modeling, drug screening, tissue engineering, and organogenesis are reviewed. Finally, this review concludes by emphasizing persistent issues and future perspectives, encouraging researchers to adopt multispheroid assembly techniques for generating advanced 3D cell models that better resemble real tissues.
Collapse
Affiliation(s)
- Tong Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haitao Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haijun Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
34
|
Tignard P, Pottin K, Geeverding A, Doulazmi M, Cabrera M, Fouquet C, Liffran M, Fouchard J, Rosello M, Albadri S, Del Bene F, Trembleau A, Breau MA. Laminin γ1-dependent basement membranes are instrumental to ensure proper olfactory placode shape, position and boundary with the brain, as well as olfactory axon development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.29.547040. [PMID: 39253416 PMCID: PMC11383033 DOI: 10.1101/2023.06.29.547040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Despite recent progress, the complex roles played by the extracellular matrix in development and disease are still far from being fully understood. Here, we took advantage of the zebrafish sly mutation which affects Laminin γ1, a major component of basement membranes, to explore its role in the development of the olfactory system. Following a detailed characterisation of Laminin distribution in the developing olfactory circuit, we analysed basement membrane integrity, olfactory placode and brain morphogenesis, and olfactory axon development in sly mutants, using a combination of immunochemistry, electron microscopy and quantitative live imaging of cell movements and axon behaviours. Our results point to an original and dual contribution of Laminin γ1-dependent basement membranes in organising the border between the olfactory placode and the adjacent brain: they maintain placode shape and position in the face of major brain morphogenetic movements, they establish a robust physical barrier between the two tissues while at the same time allowing the local entry of the sensory axons into the brain and their navigation towards the olfactory bulb. This work thus identifies key roles of Laminin γ1-dependent basement membranes in neuronal tissue morphogenesis and axon development in vivo .
Collapse
|
35
|
Lichtenberg J, Leonard CE, Sterling HR, Santos Agreda V, Hwang PY. Using Microfluidics to Align Matrix Architecture and Generate Chemokine Gradients Promotes Directional Branching in a Model of Epithelial Morphogenesis. ACS Biomater Sci Eng 2024; 10:4865-4877. [PMID: 39007451 PMCID: PMC11322918 DOI: 10.1021/acsbiomaterials.4c00245] [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: 02/05/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
The mechanical cue of fiber alignment plays a key role in the development of various tissues in the body. The ability to study the effect of these stimuli in vitro has been limited previously. Here, we present a microfluidic device capable of intrinsically generating aligned fibers using the microchannel geometry. The device also features tunable interstitial fluid flow and the ability to form a morphogen gradient. These aspects allow for the modeling of complex tissues and to differentiate cell response to different stimuli. To demonstrate the abilities of our device, we incorporated luminal epithelial cysts into our device and induced growth factor stimulation. We found the mechanical cue of fiber alignment to play a dominant role in cell elongation and the ability to form protrusions was dependent on cadherin-3. Together, this work serves as a springboard for future potential with these devices to answer questions in developmental biology and complex diseases such as cancers.
Collapse
Affiliation(s)
- Jessanne
Y. Lichtenberg
- Department
of Biomedical Engineering, Virginia Commonwealth
University, Richmond, Virginia 23220, United States
| | - Corinne E. Leonard
- Department
of Biomedical Engineering, Virginia Commonwealth
University, Richmond, Virginia 23220, United States
| | - Hazel R. Sterling
- Department
of Biomedical Engineering, Virginia Commonwealth
University, Richmond, Virginia 23220, United States
| | - Valentina Santos Agreda
- Department
of Biomedical Engineering, Virginia Commonwealth
University, Richmond, Virginia 23220, United States
| | - Priscilla Y. Hwang
- Department
of Biomedical Engineering, Virginia Commonwealth
University, Richmond, Virginia 23220, United States
- Massey
Comprehensive Cancer Center, Virginia Commonwealth
University School of Medicine, Richmond, Virginia 23298, United States
| |
Collapse
|
36
|
Aydin H, Ozcelikkale A, Acar A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater Sci Eng 2024; 10:4682-4700. [PMID: 38967485 PMCID: PMC11322920 DOI: 10.1021/acsbiomaterials.4c00445] [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: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.
Collapse
Affiliation(s)
- Hakan
Berk Aydin
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| | - Altug Ozcelikkale
- Department
of Mechanical Engineering, Middle East Technical
University, 06800, Ankara, Turkey
- Graduate
Program of Biomedical Engineering, Middle
East Technical University, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| |
Collapse
|
37
|
Rudolph EL, Chin L. Mechanobiology in Metabolic Dysfunction-Associated Steatotic Liver Disease and Obesity. Curr Issues Mol Biol 2024; 46:7134-7146. [PMID: 39057066 PMCID: PMC11276231 DOI: 10.3390/cimb46070425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
With the ongoing obesity epidemic, the prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is expected to rise and necessitates a greater understanding of how the disease proceeds from benign excess lipid in hepatocytes to liver fibrosis and eventually to liver cancer. MASLD is caused, at least in part, by hepatocytes' storage of free fatty acids (FAs) that dysfunctional adipocytes are no longer able to store, and therefore, MASLD is a disease that involves both the liver and adipose tissues. The disease progression is not only facilitated by biochemical signals, but also by mechanical cues such as the increase in stiffness often seen with fibrotic fatty livers. The change in stiffness and accumulation of excess lipid droplets impact the ability of a cell to mechanosense and mechanotranduce, which perpetuates the disease. A mechanosensitive protein that is largely unexplored and could serve as a potential therapeutic target is the intermediate filament vimentin. In this review, we briefly summarize the recent research on hepatocyte and adipocyte mechanobiology and provide a synopsis of studies on the varied, and sometimes contradictory, roles of vimentin. This review is intended to benefit and encourage future studies on hepatocyte and adipocyte mechanobiology in the context of MASLD and obesity.
Collapse
Affiliation(s)
| | - LiKang Chin
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA;
| |
Collapse
|
38
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-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: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
39
|
Naganathan SR. An emerging role for tissue plasticity in developmental precision. Biochem Soc Trans 2024; 52:987-995. [PMID: 38716859 PMCID: PMC11346420 DOI: 10.1042/bst20230173] [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: 11/08/2023] [Revised: 03/21/2024] [Accepted: 04/23/2024] [Indexed: 06/27/2024]
Abstract
Reproducible tissue morphology is a fundamental feature of embryonic development. To ensure such robustness during tissue morphogenesis, inherent noise in biological processes must be buffered. While redundant genes, parallel signaling pathways and intricate network topologies are known to reduce noise, over the last few years, mechanical properties of tissues have been shown to play a vital role. Here, taking the example of somite shape changes, I will discuss how tissues are highly plastic in their ability to change shapes leading to increased precision and reproducibility.
Collapse
Affiliation(s)
- Sundar Ram Naganathan
- Department of Biological Sciences, Tata Institute of Fundamental Research, 1, Dr. Homi Bhabha Road, Colaba, Mumbai 400005, India
| |
Collapse
|
40
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
41
|
Polsani N, Yung T, Thomas E, Phung-Rojas M, Gupta I, Denker J, Lau K, Feng X, Ibarra B, Hopyan S, Atit RP. Mesenchymal Wnts are required for morphogenetic movements of calvarial osteoblasts during apical expansion. Development 2024; 151:dev202596. [PMID: 38814743 PMCID: PMC11234264 DOI: 10.1242/dev.202596] [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: 12/08/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
Apical expansion of calvarial osteoblast progenitors from the cranial mesenchyme (CM) above the eye is integral to calvarial growth and enclosure of the brain. The cellular behaviors and signals underlying the morphogenetic process of calvarial expansion are unknown. Time-lapse light-sheet imaging of mouse embryos revealed calvarial progenitors intercalate in 3D in the CM above the eye, and exhibit protrusive and crawling activity more apically. CM cells express non-canonical Wnt/planar cell polarity (PCP) core components and calvarial osteoblasts are bidirectionally polarized. We found non-canonical ligand Wnt5a-/- mutants have less dynamic cell rearrangements and protrusive activity. Loss of CM-restricted Wntless (CM-Wls), a gene required for secretion of all Wnt ligands, led to diminished apical expansion of Osx+ calvarial osteoblasts in the frontal bone primordia in a non-cell autonomous manner without perturbing proliferation or survival. Calvarial osteoblast polarization, progressive cell elongation and enrichment for actin along the baso-apical axis were dependent on CM-Wnts. Thus, CM-Wnts regulate cellular behaviors during calvarial morphogenesis for efficient apical expansion of calvarial osteoblasts. These findings also offer potential insights into the etiologies of calvarial dysplasias.
Collapse
Affiliation(s)
- Nikaya Polsani
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Theodora Yung
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Evan Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Melissa Phung-Rojas
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Isha Gupta
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Julie Denker
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Kimberly Lau
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiaotian Feng
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Beatriz Ibarra
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Orthopedics, The Hospital for Sick Children and Departments of Molecular Genetics and Surgery, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Radhika P. Atit
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Dermatology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Genetics and Genome Sciences, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
42
|
Dibus M, Joshi O, Ivaska J. Novel tools to study cell-ECM interactions, cell adhesion dynamics and migration. Curr Opin Cell Biol 2024; 88:102355. [PMID: 38631101 DOI: 10.1016/j.ceb.2024.102355] [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: 12/01/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
Integrin-mediated cell adhesion is essential for cell migration, mechanotransduction and tissue integrity. In vivo, these processes are regulated by complex physicochemical signals from the extracellular matrix (ECM). These nuanced cues, including molecular composition, rigidity and topology, call for sophisticated systems to faithfully explore cell behaviour. Here, we discuss recent methodological advances in cell-ECM adhesion research and compile a toolbox of techniques that we expect to shape this field in future. We outline methodological breakthroughs facilitating the transition from rigid 2D substrates to more complex and dynamic 3D systems, as well as advances in super-resolution imaging for an in-depth understanding of adhesion nanostructure. Selected methods are exemplified with relevant biological findings to underscore their applicability in cell adhesion research. We expect this new "toolbox" of methods will allow for a closer approximation of in vitro experimental setups to in vivo conditions, providing deeper insights into physiological and pathophysiological processes associated with cell-ECM adhesion.
Collapse
Affiliation(s)
- Michal Dibus
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Omkar Joshi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, FI-20520 Turku, Finland; Western Finnish Cancer Center (FICAN West), University of Turku, FI-20520 Turku, Finland; Foundation for the Finnish Cancer Institute, Tukholmankatu 8, FI-00014 Helsinki, Finland.
| |
Collapse
|
43
|
Lunova M, Jirsa M, Dejneka A, Sullivan GJ, Lunov O. Mechanical regulation of mitochondrial morphodynamics in cancer cells by extracellular microenvironment. BIOMATERIALS AND BIOSYSTEMS 2024; 14:100093. [PMID: 38585282 PMCID: PMC10992729 DOI: 10.1016/j.bbiosy.2024.100093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/05/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024] Open
Abstract
Recently, it has been recognized that physical abnormalities (e.g. elevated solid stress, elevated interstitial fluid pressure, increased stiffness) are associated with tumor progression and development. Additionally, these mechanical forces originating from tumor cell environment through mechanotransduction pathways can affect metabolism. On the other hand, mitochondria are well-known as bioenergetic, biosynthetic, and signaling organelles crucial for sensing stress and facilitating cellular adaptation to the environment and physical stimuli. Disruptions in mitochondrial dynamics and function have been found to play a role in the initiation and advancement of cancer. Consequently, it is logical to hypothesize that mitochondria dynamics subjected to physical cues may play a pivotal role in mediating tumorigenesis. Recently mitochondrial biogenesis and turnover, fission and fusion dynamics was linked to mechanotransduction in cancer. However, how cancer cell mechanics and mitochondria functions are connected, still remain poorly understood. Here, we discuss recent studies that link mechanical stimuli exerted by the tumor cell environment and mitochondria dynamics and functions. This interplay between mechanics and mitochondria functions may shed light on how mitochondria regulate tumorigenesis.
Collapse
Affiliation(s)
- Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18200, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), Prague 14021, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague 14021, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18200, Czech Republic
| | | | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18200, Czech Republic
| |
Collapse
|
44
|
Sun Q, Pan X, Wang P, Wei Q. Synergistic Influence of Fibrous Pattern Orientation and Modulus on Cellular Mechanoresponse. NANO LETTERS 2024; 24:6376-6385. [PMID: 38743504 DOI: 10.1021/acs.nanolett.4c01352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The fibrous extracellular matrix (ECM) is vital for tissue regeneration and impacts implanted device treatments. Previous research on fibrous biomaterials shows varying cellular reactions to surface orientation, often due to unclear interactions between surface topography and substrate elasticity. Our study addresses this gap by achieving the rapid creation of hydrogels with diverse fibrous topographies and varying substrate moduli through a surface printing strategy. Cells exhibit heightened traction force on nanopatterned soft hydrogels, particularly with randomly distributed patterns compared with regular soft hydrogels. Meanwhile, on stiff hydrogels featuring an aligned topography, optimal cellular mechanosensing is observed compared to random topography. Mechanistic investigations highlight that cellular force-sensing and adhesion are influenced by the interplay of pattern deformability and focal adhesion orientation, subsequently mediating stem cell differentiation. Our findings highlight the importance of combining substrate modulus and topography to guide cellular behavior in designing advanced tissue engineering biomaterials.
Collapse
Affiliation(s)
- Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaokai Pan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
45
|
Sun Z, Chen Z, Yin M, Wu X, Guo B, Cheng X, Quan R, Sun Y, Zhang Q, Fan Y, Jin C, Yin Y, Hou X, Liu W, Shu M, Xue X, Shi Y, Chen B, Xiao Z, Dai J, Zhao Y. Harnessing developmental dynamics of spinal cord extracellular matrix improves regenerative potential of spinal cord organoids. Cell Stem Cell 2024; 31:772-787.e11. [PMID: 38565140 DOI: 10.1016/j.stem.2024.03.007] [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: 10/17/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Neonatal spinal cord tissues exhibit remarkable regenerative capabilities as compared to adult spinal cord tissues after injury, but the role of extracellular matrix (ECM) in this process has remained elusive. Here, we found that early developmental spinal cord had higher levels of ECM proteins associated with neural development and axon growth, but fewer inhibitory proteoglycans, compared to those of adult spinal cord. Decellularized spinal cord ECM from neonatal (DNSCM) and adult (DASCM) rabbits preserved these differences. DNSCM promoted proliferation, migration, and neuronal differentiation of neural progenitor cells (NPCs) and facilitated axonal outgrowth and regeneration of spinal cord organoids more effectively than DASCM. Pleiotrophin (PTN) and Tenascin (TNC) in DNSCM were identified as contributors to these abilities. Furthermore, DNSCM demonstrated superior performance as a delivery vehicle for NPCs and organoids in spinal cord injury (SCI) models. This suggests that ECM cues from early development stages might significantly contribute to the prominent regeneration ability in spinal cord.
Collapse
Affiliation(s)
- Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenni Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaokang Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui Quan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Jin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Muya Shu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
46
|
Olaizola-Rodrigo C, Castro-Abril H, Perisé-Badía I, Pancorbo L, Ochoa I, Monge R, Oliván S. Reducing Inert Materials for Optimal Cell-Cell and Cell-Matrix Interactions within Microphysiological Systems. Biomimetics (Basel) 2024; 9:262. [PMID: 38786472 PMCID: PMC11118140 DOI: 10.3390/biomimetics9050262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
In the pursuit of achieving a more realistic in vitro simulation of human biological tissues, microfluidics has emerged as a promising technology. Organ-on-a-chip (OoC) devices, a product of this technology, contain miniature tissues within microfluidic chips, aiming to closely mimic the in vivo environment. However, a notable drawback is the presence of inert material between compartments, hindering complete contact between biological tissues. Current membranes, often made of PDMS or plastic materials, prevent full interaction between cell types and nutrients. Furthermore, their non-physiological mechanical properties and composition may induce unexpected cell responses. Therefore, it is essential to minimize the contact area between cells and the inert materials while simultaneously maximizing the direct contact between cells and matrices in different compartments. The main objective of this work is to minimize inert materials within the microfluidic chip while preserving proper cellular distribution. Two microfluidic devices were designed, each with a specific focus on maximizing direct cell-matrix or cell-cell interactions. The first chip, designed to increase direct cell-cell interactions, incorporates a nylon mesh with regular pores of 150 microns. The second chip minimizes interference from inert materials, thereby aiming to increase direct cell-matrix contact. It features an inert membrane with optimized macropores of 1 mm of diameter for collagen hydrogel deposition. Biological validation of both devices has been conducted through the implementation of cell migration and cell-to-cell interaction assays, as well as the development of epithelia, from isolated cells or spheroids. This endeavor contributes to the advancement of microfluidic technology, aimed at enhancing the precision and biological relevance of in vitro simulations in pursuit of more biomimetic models.
Collapse
Affiliation(s)
- Claudia Olaizola-Rodrigo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Héctor Castro-Abril
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Laboratorio de Biomiméticos, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Ismael Perisé-Badía
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lara Pancorbo
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Monge
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
47
|
Heyn JCJ, Rädler JO, Falcke M. Mesenchymal cell migration on one-dimensional micropatterns. Front Cell Dev Biol 2024; 12:1352279. [PMID: 38694822 PMCID: PMC11062138 DOI: 10.3389/fcell.2024.1352279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/29/2024] [Indexed: 05/04/2024] Open
Abstract
Quantitative studies of mesenchymal cell motion are important to elucidate cytoskeleton function and mechanisms of cell migration. To this end, confinement of cell motion to one dimension (1D) significantly simplifies the problem of cell shape in experimental and theoretical investigations. Here we review 1D migration assays employing micro-fabricated lanes and reflect on the advantages of such platforms. Data are analyzed using biophysical models of cell migration that reproduce the rich scenario of morphodynamic behavior found in 1D. We describe basic model assumptions and model behavior. It appears that mechanical models explain the occurrence of universal relations conserved across different cell lines such as the adhesion-velocity relation and the universal correlation between speed and persistence (UCSP). We highlight the unique opportunity of reproducible and standardized 1D assays to validate theory based on statistical measures from large data of trajectories and discuss the potential of experimental settings embedding controlled perturbations to probe response in migratory behavior.
Collapse
Affiliation(s)
- Johannes C. J. Heyn
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Joachim O. Rädler
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Physics, Humboldt University, Berlin, Germany
| |
Collapse
|
48
|
Mun S, Lee HJ, Kim P. Rebuilding the microenvironment of primary tumors in humans: a focus on stroma. Exp Mol Med 2024; 56:527-548. [PMID: 38443595 PMCID: PMC10984944 DOI: 10.1038/s12276-024-01191-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/05/2023] [Accepted: 12/29/2023] [Indexed: 03/07/2024] Open
Abstract
Conventional tumor models have critical shortcomings in that they lack the complexity of the human stroma. The heterogeneous stroma is a central compartment of the tumor microenvironment (TME) that must be addressed in cancer research and precision medicine. To fully model the human tumor stroma, the deconstruction and reconstruction of tumor tissues have been suggested as new approaches for in vitro tumor modeling. In this review, we summarize the heterogeneity of tumor-associated stromal cells and general deconstruction approaches used to isolate patient-specific stromal cells from tumor tissue; we also address the effect of the deconstruction procedure on the characteristics of primary cells. Finally, perspectives on the future of reconstructed tumor models are discussed, with an emphasis on the essential prerequisites for developing authentic humanized tumor models.
Collapse
Affiliation(s)
- Siwon Mun
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, South Korea
| | - Hyun Jin Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, South Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, South Korea.
- Institute for Health Science and Technology, KAIST, Daejeon, 34141, South Korea.
| |
Collapse
|
49
|
Ferreira B, Barros AS, Leite-Pereira C, Viegas J, das Neves J, Nunes R, Sarmento B. Trends in 3D models of inflammatory bowel disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167042. [PMID: 38296115 DOI: 10.1016/j.bbadis.2024.167042] [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: 10/23/2023] [Revised: 12/11/2023] [Accepted: 01/25/2024] [Indexed: 02/20/2024]
Abstract
Inflammatory bowel disease (IBD) encompasses a set of chronic inflammatory conditions, namely Crohn's disease and ulcerative colitis. Despite all advances in the management of IBD, a definitive cure is not available, largely due to a lack of a holistic understanding of its etiology and pathophysiology. Several in vitro, in vivo, and ex vivo models have been developed over the past few decades in order to abbreviate remaining gaps. The establishment of reliable and predictable in vitro intestinal inflammation models may indeed provide valuable tools to expedite and validate the development of therapies for IBD. Three-dimensional (3D) models provide a more accurate representation of the different layers of the intestine, contributing to a stronger impact on drug screening and research on intestinal inflammation, and bridging the gap between in vitro and in vivo research. This work provides a critical overview on the state-of-the-art on existing 3D models of intestinal inflammation and discusses the remaining challenges, providing insights on possible pathways towards achieving IBD mimetic models. We also address some of the main challenges faced by implementing cell culture models in IBD research while bearing in mind clinical translational aspects.
Collapse
Affiliation(s)
- Bárbara Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Andreia S Barros
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Catarina Leite-Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Juliana Viegas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - José das Neves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Rute Nunes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
50
|
Hamel KM, Frazier TP, Williams C, Duplessis T, Rowan BG, Gimble JM, Sanchez CG. Adipose Tissue in Breast Cancer Microphysiological Models to Capture Human Diversity in Preclinical Models. Int J Mol Sci 2024; 25:2728. [PMID: 38473978 PMCID: PMC10931959 DOI: 10.3390/ijms25052728] [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: 02/01/2024] [Revised: 02/23/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Female breast cancer accounts for 15.2% of all new cancer cases in the United States, with a continuing increase in incidence despite efforts to discover new targeted therapies. With an approximate failure rate of 85% for therapies in the early phases of clinical trials, there is a need for more translatable, new preclinical in vitro models that include cellular heterogeneity, extracellular matrix, and human-derived biomaterials. Specifically, adipose tissue and its resident cell populations have been identified as necessary attributes for current preclinical models. Adipose-derived stromal/stem cells (ASCs) and mature adipocytes are a normal part of the breast tissue composition and not only contribute to normal breast physiology but also play a significant role in breast cancer pathophysiology. Given the recognized pro-tumorigenic role of adipocytes in tumor progression, there remains a need to enhance the complexity of current models and account for the contribution of the components that exist within the adipose stromal environment to breast tumorigenesis. This review article captures the current landscape of preclinical breast cancer models with a focus on breast cancer microphysiological system (MPS) models and their counterpart patient-derived xenograft (PDX) models to capture patient diversity as they relate to adipose tissue.
Collapse
Affiliation(s)
- Katie M. Hamel
- Obatala Sciences, Inc., New Orleans, LA 70148, USA; (K.M.H.); (T.P.F.); (J.M.G.)
| | - Trivia P. Frazier
- Obatala Sciences, Inc., New Orleans, LA 70148, USA; (K.M.H.); (T.P.F.); (J.M.G.)
| | - Christopher Williams
- Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | | | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Jeffrey M. Gimble
- Obatala Sciences, Inc., New Orleans, LA 70148, USA; (K.M.H.); (T.P.F.); (J.M.G.)
| | - Cecilia G. Sanchez
- Obatala Sciences, Inc., New Orleans, LA 70148, USA; (K.M.H.); (T.P.F.); (J.M.G.)
| |
Collapse
|