1
|
Houtekamer RM, van der Net MC, Vliem MJ, Noordzij TEJC, van Uden L, van Es RM, Sim JY, Deguchi E, Terai K, Hopcroft MA, Vos HR, Pruitt BL, Matsuda M, Pannekoek WJ, Gloerich M. E-cadherin mechanotransduction activates EGFR-ERK signaling in epithelial monolayers by inducing ADAM-mediated ligand shedding. Sci Signal 2025; 18:eadr7926. [PMID: 40359261 DOI: 10.1126/scisignal.adr7926] [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: 07/16/2024] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
The behavior of cells is governed by signals originating from their local environment, including mechanical forces exerted on the cells. Forces are transduced by mechanosensitive proteins, which can impinge on signaling cascades that are also activated by growth factors. We investigated the cross-talk between mechanical and biochemical signals in the regulation of intracellular signaling networks in epithelial monolayers. Phosphoproteomic and transcriptomic analyses on epithelial monolayers subjected to mechanical strain revealed the activation of extracellular signal-regulated kinase (ERK) downstream of the epidermal growth factor receptor (EGFR) as a predominant strain-induced signaling event. Strain-induced EGFR-ERK signaling depended on mechanosensitive E-cadherin adhesions. Proximity labeling showed that the metalloproteinase ADAM17, an enzyme that mediates shedding of soluble EGFR ligands, was closely associated with E-cadherin. A probe that we developed to monitor ADAM-mediated shedding demonstrated that mechanical strain induced ADAM activation. Mechanically induced ADAM activation was essential for mechanosensitive, E-cadherin-dependent EGFR-ERK signaling. Together, our data demonstrate that mechanical strain transduced by E-cadherin adhesion triggers the shedding of EGFR ligands that stimulate downstream ERK activity. Our findings illustrate how mechanical signals and biochemical ligands can operate within a linear signaling cascade.
Collapse
Affiliation(s)
- Ronja M Houtekamer
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Mirjam C van der Net
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Marjolein J Vliem
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Tomas E J C Noordzij
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Lisa van Uden
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Robert M van Es
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Joo Yong Sim
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Eriko Deguchi
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenta Terai
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Matthew A Hopcroft
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Harmjan R Vos
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Beth L Pruitt
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Willem-Jan Pannekoek
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Martijn Gloerich
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| |
Collapse
|
2
|
Mishchenko O, Volchykhina K, Maksymov D, Manukhina O, Pogorielov M, Pavlenko M, Iatsunskyi I. Advanced Strategies for Enhancing the Biocompatibility and Antibacterial Properties of Implantable Structures. MATERIALS (BASEL, SWITZERLAND) 2025; 18:822. [PMID: 40004345 PMCID: PMC11857362 DOI: 10.3390/ma18040822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
This review explores the latest advancements in enhancing the biocompatibility and antibacterial properties of implantable structures, with a focus on titanium (Ti) and its alloys. Titanium implants, widely used in dental and orthopedic applications, demonstrate excellent mechanical strength and biocompatibility, yet face challenges such as peri-implantitis, a bacterial infection that can lead to implant failure. To address these issues, both passive and active surface modification strategies have been developed. Passive modifications, such as altering surface texture and chemistry, aim to prevent bacterial adhesion, while active approaches incorporate antimicrobial agents for sustained infection control. Nanotechnology has emerged as a transformative tool, enabling the creation of nanoscale materials and coatings like TiO2 and ZnO that promote osseointegration and inhibit biofilm formation. Techniques such as plasma spraying, ion implantation, and plasma electrolytic oxidation (PEO) show promising results in improving implant integration and durability. Despite significant progress, further research is needed to refine these technologies, optimize surface properties, and address the clinical challenges associated with implant longevity and safety. This review highlights the intersection of surface engineering, nanotechnology, and biomedical innovation, paving the way for the next generation of implantable devices.
Collapse
Affiliation(s)
- Oleg Mishchenko
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Kristina Volchykhina
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Denis Maksymov
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Olesia Manukhina
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Maksym Pogorielov
- Insitute of Atomic Physics and Spectroscopy, University of Latvia, 3 Jelgavas Str., LV-1004 Riga, Latvia;
| | - Mykola Pavlenko
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej Str. 3, 61-614 Poznan, Poland;
| | - Igor Iatsunskyi
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej Str. 3, 61-614 Poznan, Poland;
| |
Collapse
|
3
|
Wu P, Sawaki S, Yamauchi K, Yokota K, Hakamada M, Mabuchi M. Long range juxtacrine signalling through cadherin for collective cell orientation. Acta Biomater 2024:S1742-7061(24)00627-5. [PMID: 39454932 DOI: 10.1016/j.actbio.2024.10.035] [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: 07/02/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Many life phenomena, such as development, morphogenesis, tissue remodelling, and wound healing, are often driven by orderly and directional migration of collective cells. However, when cells are randomly oriented or localized disorder exists in orderly oriented collective cells, cell migration cannot occur in an orderly manner although various motion modes such as global rotation and local swirling and/or various motion patterns such as radial pattern and chiral pattern often occur. Therefore, it is important to control cell orientation to ensure the orderly migration of collective cells. Here, we show that it is not force transmission, but juxtacrine signalling through cadherin that plays a critical role in the orientation of collective cells. Surprisingly, juxtacrine signalling for cell orientation reached cells on a plastic dish that were not directly subjected to mechanical stimulation, up to 7 mm away from the actuator. The present study suggests that even weak mechanical stimulation is transmitted in a long range without force transmission through juxtacrine signalling. The long range juxtacrine signalling might play an important role in various life phenomena. STATEMENT OF SIGNIFICANCE: Juxtacrine signalling is direct cell-cell contact-dependent signalling, which plays a crucial role in cell behaviors such as mechanosensing, mechanotransduction and collective cell behaviors, however, there is not enough understanding about juxtacrine signalling. The present study has demonstrated that juxtacrine signalling for collective cell orientation is transmitted over a long range through cadherin. To the best of our knowledge, this is the first report of long range juxtacrine signalling. This finding may lead to the elucidation of various life phenomena such as development, morphogenesis, tissue remodelling, and wound healing.
Collapse
Affiliation(s)
- Peizheng Wu
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan.
| | - Shogo Sawaki
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Kei Yamauchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Kazuki Yokota
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Masataka Hakamada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Mamoru Mabuchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| |
Collapse
|
4
|
Liu Y, Murazzi I, Fuller AM, Pan H, Irizarry-Negron VM, Devine A, Katti R, Skuli N, Ciotti GE, Pak K, Pack MA, Simon MC, Weber K, Cooper K, Eisinger-Mathason TK. Sarcoma Cells Secrete Hypoxia-Modified Collagen VI to Weaken the Lung Endothelial Barrier and Promote Metastasis. Cancer Res 2024; 84:977-993. [PMID: 38335278 PMCID: PMC10984776 DOI: 10.1158/0008-5472.can-23-0910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 12/21/2023] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Intratumoral hypoxia correlates with metastasis and poor survival in patients with sarcoma. Using an impedance sensing assay and a zebrafish intravital microinjection model, we demonstrated here that the hypoxia-inducible collagen-modifying enzyme lysyl hydroxylase PLOD2 and its substrate collagen type VI (COLVI) weaken the lung endothelial barrier and promote transendothelial migration. Mechanistically, hypoxia-induced PLOD2 in sarcoma cells modified COLVI, which was then secreted into the vasculature. Upon reaching the apical surface of lung endothelial cells, modified COLVI from tumor cells activated integrin β1 (ITGβ1). Furthermore, activated ITGβ1 colocalized with Kindlin2, initiating their interaction with F-actin and prompting its polymerization. Polymerized F-actin disrupted endothelial adherens junctions and induced barrier dysfunction. Consistently, modified and secreted COLVI was required for the late stages of lung metastasis in vivo. Analysis of patient gene expression and survival data from The Cancer Genome Atlas (TCGA) revealed an association between the expression of both PLOD2 and COLVI and patient survival. Furthermore, high levels of COLVI were detected in surgically resected sarcoma metastases from patient lungs and in the blood of tumor-bearing mice. Together, these data identify a mechanism of sarcoma lung metastasis, revealing opportunities for therapeutic intervention. SIGNIFICANCE Collagen type VI modified by hypoxia-induced PLOD2 is secreted by sarcoma cells and binds to integrin β1 on endothelial cells to induce barrier dysfunction, which promotes sarcoma vascular dissemination and metastasis.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ashley M. Fuller
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Hehai Pan
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie M Irizarry-Negron
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Ann Devine
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Rohan Katti
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas Skuli
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- Department of Cell and Developmental Biology
- University of Pennsylvania, Philadelphia, PA, USA
| | - Gabrielle E. Ciotti
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Koreana Pak
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A. Pack
- Perelman School of Medicine
- Department of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - M. Celeste Simon
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- Department of Cell and Developmental Biology
- University of Pennsylvania, Philadelphia, PA, USA
| | - Kristy Weber
- Penn Sarcoma Program
- Perelman School of Medicine
- Department of Orthopedic Surgery
- University of Pennsylvania, Philadelphia, PA, USA
| | - Kumarasen Cooper
- Department of Pathology & Laboratory Medicine
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| | - T.S. Karin Eisinger-Mathason
- Department of Pathology & Laboratory Medicine
- Penn Sarcoma Program
- Abramson Family Cancer Research Institute
- Perelman School of Medicine
- University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Ebrahim T, Ebrahim AS, Kandouz M. Diversity of Intercellular Communication Modes: A Cancer Biology Perspective. Cells 2024; 13:495. [PMID: 38534339 PMCID: PMC10969453 DOI: 10.3390/cells13060495] [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: 01/05/2024] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
From the moment a cell is on the path to malignant transformation, its interaction with other cells from the microenvironment becomes altered. The flow of molecular information is at the heart of the cellular and systemic fate in tumors, and various processes participate in conveying key molecular information from or to certain cancer cells. For instance, the loss of tight junction molecules is part of the signal sent to cancer cells so that they are no longer bound to the primary tumors and are thus free to travel and metastasize. Upon the targeting of a single cell by a therapeutic drug, gap junctions are able to communicate death information to by-standing cells. The discovery of the importance of novel modes of cell-cell communication such as different types of extracellular vesicles or tunneling nanotubes is changing the way scientists look at these processes. However, are they all actively involved in different contexts at the same time or are they recruited to fulfill specific tasks? What does the multiplicity of modes mean for the overall progression of the disease? Here, we extend an open invitation to think about the overall significance of these questions, rather than engage in an elusive attempt at a systematic repertory of the mechanisms at play.
Collapse
Affiliation(s)
- Thanzeela Ebrahim
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48202, USA
| |
Collapse
|
6
|
Ahmed TA, Ahmed SM, Elkhenany H, El-Desouky MA, Magdeldin S, Osama A, Anwar AM, Mohamed IK, Abdelgawad ME, Hanna DH, El-Badri N. The cross talk between type II diabetic microenvironment and the regenerative capacities of human adipose tissue-derived pericytes: a promising cell therapy. Stem Cell Res Ther 2024; 15:36. [PMID: 38331889 PMCID: PMC10854071 DOI: 10.1186/s13287-024-03643-1] [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: 08/23/2023] [Accepted: 01/21/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Pericytes (PCs) are multipotent contractile cells that wrap around the endothelial cells (ECs) to maintain the blood vessel's functionality and integrity. The hyperglycemia associated with Type 2 diabetes mellitus (T2DM) was shown to impair the function of PCs and increase the risk of diabetes complications. In this study, we aimed to investigate the deleterious effect of the diabetic microenvironment on the regenerative capacities of human PCs. METHODS PCs isolated from human adipose tissue were cultured in the presence or absence of serum collected from diabetic patients. The functionality of PCs was analyzed after 6, 14, and 30 days. RESULTS Microscopic examination of PCs cultured in DS (DS-PCs) showed increased aggregate formation and altered surface topography with hyperbolic invaginations. Compared to PCs cultured in normal serum (NS-PCs), DS-PCs showed more fragmented mitochondria and thicker nuclear membrane. DS caused impaired angiogenic differentiation of PCs as confirmed by tube formation, decreased VEGF-A and IGF-1 gene expression, upregulated TSP1, PF4, actin-related protein 2/3 complex, and downregulated COL21A1 protein expression. These cells suffered more pronounced apoptosis and showed higher expression of Clic4, apoptosis facilitator BCl-2-like protein, serine/threonine protein phosphatase, and caspase-7 proteins. DS-PCs showed dysregulated DNA repair genes CDKN1A, SIRT1, XRCC5 TERF2, and upregulation of the pro-inflammatory genes ICAM1, IL-6, and TNF-α. Further, DS-treated cells also showed disruption in the expression of the focal adhesion and binding proteins TSP1, TGF-β, fibronectin, and PCDH7. Interestingly, DS-PCs showed resistance mechanisms upon exposure to diabetic microenvironment by maintaining the intracellular reactive oxygen species (ROS) level and upregulation of extracellular matrix (ECM) organizing proteins as vinculin, IQGAP1, and tubulin beta chain. CONCLUSION These data showed that the diabetic microenvironment exert a deleterious effect on the regenerative capacities of human adipose tissue-derived PCs, and may thus have possible implications on the vascular complications of T2DM. Nevertheless, PCs have shown remarkable protective mechanisms when initially exposed to DS and thus they could provide a promising cellular therapy for T2DM.
Collapse
Affiliation(s)
- Toka A Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Sara M Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Hoda Elkhenany
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22785, Egypt
| | - Mohamed A El-Desouky
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Basic Research Department, Children's Cancer Hospital, Cairo, 57357, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Aya Osama
- Proteomics and Metabolomics Research Program, Basic Research Department, Children's Cancer Hospital, Cairo, 57357, Egypt
| | - Ali Mostafa Anwar
- Proteomics and Metabolomics Research Program, Basic Research Department, Children's Cancer Hospital, Cairo, 57357, Egypt
| | - Ihab K Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohamed Essameldin Abdelgawad
- Biochemistry and Molecular Biotechnology Division, Chemistry Department, Faculty of Science, Innovative Cellular Microenvironment Optimization Platform (ICMOP), Precision Therapy Unit, Helwan University, Cairo, Egypt
- The Egyptian Network of Bioinformatics "BioNetMasr", Cairo, Egypt
| | - Demiana H Hanna
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt.
| |
Collapse
|
7
|
Chirasani VR, Khan MAI, Malavade JN, Dokholyan NV, Hoffman BD, Campbell SL. Molecular basis and cellular functions of vinculin-actin directional catch bonding. Nat Commun 2023; 14:8300. [PMID: 38097542 PMCID: PMC10721916 DOI: 10.1038/s41467-023-43779-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
The ability of cells and tissues to respond differentially to mechanical forces applied in distinct directions is mediated by the ability of load-bearing proteins to preferentially maintain physical linkages in certain directions. However, the molecular basis and biological consequences of directional force-sensitive binding remain unclear. Vinculin (Vcn) is a load-bearing linker protein that exhibits directional catch bonding due to interactions between the Vcn tail domain (Vt) and filamentous (F)-actin. We developed a computational approach to predict Vcn residues involved in directional catch bonding and produced a set of associated Vcn variants with unaltered Vt structure, actin binding, or phospholipid interactions. Incorporation of the variants did not affect Vcn activation but reduced Vcn loading and altered exchange dynamics, consistent with the loss of directional catch bonding. Expression of Vcn variants perturbed the coordination of subcellular structures and cell migration, establishing key cellular functions for Vcn directional catch bonding.
Collapse
Affiliation(s)
- Venkat R Chirasani
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohammad Ashhar I Khan
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juilee N Malavade
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Chemistry, Penn State College of Medicine, Hershey, PA, USA.
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Cell Biology, Duke University, Durham, NC, USA.
| | - Sharon L Campbell
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
Valdivia A, Avalos AM, Leyton L. Thy-1 (CD90)-regulated cell adhesion and migration of mesenchymal cells: insights into adhesomes, mechanical forces, and signaling pathways. Front Cell Dev Biol 2023; 11:1221306. [PMID: 38099295 PMCID: PMC10720913 DOI: 10.3389/fcell.2023.1221306] [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: 05/12/2023] [Accepted: 09/25/2023] [Indexed: 12/17/2023] Open
Abstract
Cell adhesion and migration depend on the assembly and disassembly of adhesive structures known as focal adhesions. Cells adhere to the extracellular matrix (ECM) and form these structures via receptors, such as integrins and syndecans, which initiate signal transduction pathways that bridge the ECM to the cytoskeleton, thus governing adhesion and migration processes. Integrins bind to the ECM and soluble or cell surface ligands to form integrin adhesion complexes (IAC), whose composition depends on the cellular context and cell type. Proteomic analyses of these IACs led to the curation of the term adhesome, which is a complex molecular network containing hundreds of proteins involved in signaling, adhesion, and cell movement. One of the hallmarks of these IACs is to sense mechanical cues that arise due to ECM rigidity, as well as the tension exerted by cell-cell interactions, and transduce this force by modifying the actin cytoskeleton to regulate cell migration. Among the integrin/syndecan cell surface ligands, we have described Thy-1 (CD90), a GPI-anchored protein that possesses binding domains for each of these receptors and, upon engaging them, stimulates cell adhesion and migration. In this review, we examine what is currently known about adhesomes, revise how mechanical forces have changed our view on the regulation of cell migration, and, in this context, discuss how we have contributed to the understanding of signaling mechanisms that control cell adhesion and migration.
Collapse
Affiliation(s)
- Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
9
|
Luo P, Huang R, Wu Y, Liu X, Shan Z, Gong L, Deng S, Liu H, Fang J, Wu S, Wu X, Liu Q, Chen Z, Yeung KW, Qiao W, Chen S, Chen Z. Tailoring the multiscale mechanics of tunable decellularized extracellular matrix (dECM) for wound healing through immunomodulation. Bioact Mater 2023; 28:95-111. [PMID: 37250862 PMCID: PMC10209339 DOI: 10.1016/j.bioactmat.2023.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/31/2023] Open
Abstract
With the discovery of the pivotal role of macrophages in tissue regeneration through shaping the tissue immune microenvironment, various immunomodulatory strategies have been proposed to modify traditional biomaterials. Decellularized extracellular matrix (dECM) has been extensively used in the clinical treatment of tissue injury due to its favorable biocompatibility and similarity to the native tissue environment. However, most reported decellularization protocols may cause damage to the native structure of dECM, which undermines its inherent advantages and potential clinical applications. Here, we introduce a mechanically tunable dECM prepared by optimizing the freeze-thaw cycles. We demonstrated that the alteration in micromechanical properties of dECM resulting from the cyclic freeze-thaw process contributes to distinct macrophage-mediated host immune responses to the materials, which are recently recognized to play a pivotal role in determining the outcome of tissue regeneration. Our sequencing data further revealed that the immunomodulatory effect of dECM was induced via the mechnotrasduction pathways in macrophages. Next, we tested the dECM in a rat skin injury model and found an enhanced micromechanical property of dECM achieved with three freeze-thaw cycles significantly promoted the M2 polarization of macrophages, leading to superior wound healing. These findings suggest that the immunomodulatory property of dECM can be efficiently manipulated by tailoring its inherent micromechanical properties during the decellularization process. Therefore, our mechanics-immunomodulation-based strategy provides new insights into the development of advanced biomaterials for wound healing.
Collapse
Affiliation(s)
- Pu Luo
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Ruoxuan Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - You Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Xingchen Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zhengjie Shan
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Li Gong
- Instrumental Analysis Research Center, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shudan Deng
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Haiwen Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Jinghan Fang
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518058, China
| | - Shiyu Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Xiayi Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Quan Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zetao Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Kelvin W.K. Yeung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518058, China
| | - Wei Qiao
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Shoucheng Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zhuofan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| |
Collapse
|
10
|
Lu S, Lu T, Zhang J, Gan L, Wu X, Han D, Zhang K, Xu C, Liu S, Qin W, Yang F, Wen W. CD248 promotes migration and metastasis of osteosarcoma through ITGB1-mediated FAK-paxillin pathway activation. BMC Cancer 2023; 23:290. [PMID: 36997926 PMCID: PMC10061858 DOI: 10.1186/s12885-023-10731-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/13/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common malignant bone tumor with a high incidence in children and adolescents. Frequent tumor metastasis and high postoperative recurrence are the most common challenges in OS. However, detailed mechanism is largely unknown. METHODS We examined the expression of CD248 in OS tissue microarrays by immunohistochemistry (IHC) staining. We studied the biological function of CD248 in cell proliferation, invasion and migration of OS cells by CCK8 assay, transwell and wound healing assay. We also studied its function in the metastasis of OS in vivo. At last, we explored the potential mechanism how CD248 promotes OS metastasis by using RNA-seq, western blot, immunofluorescence staining and co-immunoprecipitation using CD248 knockdown OS cells. RESULTS CD248 was highly expressed in OS tissues and its high expression was correlated with pulmonary metastasis of OS. Knockdown of CD248 in OS cells significantly inhibited cell migration, invasion and metastasis, while had no obvious effect on cell proliferation. Lung metastasis in nude mice was significantly inhibited when CD248 was knocked down. Mechanistically, we found that CD248 could promote the interaction between ITGB1 and extracellular matrix (ECM) proteins like CYR61 and FN, which activated the FAK-paxillin pathway to promote the formation of focal adhesion and metastasis of OS. CONCLUSION Our data showed that high CD248 expression is correlated with the metastatic potential of OS. CD248 may promote migration and metastasis through enhancing the interaction between ITGB1 and certain ECM proteins. Therefore, CD248 is a potential marker for diagnosis and effective target for the treatment of metastatic OS.
Collapse
Affiliation(s)
- Shiqi Lu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Tong Lu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jiayu Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Lunbiao Gan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Xinjie Wu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Fa Yang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Weihong Wen
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
| |
Collapse
|
11
|
Abstract
The endothelium is a dynamic, semipermeable layer lining all blood vessels, regulating blood vessel formation and barrier function. Proper composition and function of the endothelial barrier are required for fluid homeostasis, and clinical conditions characterized by barrier disruption are associated with severe morbidity and high mortality rates. Endothelial barrier properties are regulated by cell-cell junctions and intracellular signaling pathways governing the cytoskeleton, but recent insights indicate an increasingly important role for integrin-mediated cell-matrix adhesion and signaling in endothelial barrier regulation. Here, we discuss diseases characterized by endothelial barrier disruption, and provide an overview of the composition of endothelial cell-matrix adhesion complexes and associated signaling pathways, their crosstalk with cell-cell junctions, and with other receptors. We further present recent insights into the role of cell-matrix adhesions in the developing and mature/adult endothelium of various vascular beds, and discuss how the dynamic regulation and turnover of cell-matrix adhesions regulates endothelial barrier function in (patho)physiological conditions like angiogenesis, inflammation and in response to hemodynamic stress. Finally, as clinical conditions associated with vascular leak still lack direct treatment, we focus on how understanding of endothelial cell-matrix adhesion may provide novel targets for treatment, and discuss current translational challenges and future perspectives.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Pulmonology, Amsterdam University Medical Center, the Netherlands (J.A.)
| | - Coert Margadant
- Department of Medical Oncology, Amsterdam University Medical Center, the NetherlandsInstitute of Biology, Leiden University, the Netherlands (C.M.)
| |
Collapse
|
12
|
Li X, McLain C, Samuel MS, Olson MF, Radice GL. Actomyosin-mediated cellular tension promotes Yap nuclear translocation and myocardial proliferation through α5 integrin signaling. Development 2023; 150:dev201013. [PMID: 36621002 PMCID: PMC10110499 DOI: 10.1242/dev.201013] [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: 06/09/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023]
Abstract
The cardiomyocyte phenotypic switch from a proliferative to terminally differentiated state results in the loss of regenerative potential of the mammalian heart shortly after birth. Nonmuscle myosin IIB (NM IIB)-mediated actomyosin contractility regulates cardiomyocyte cytokinesis in the embryonic heart, and NM IIB levels decline after birth, suggesting a role for cellular tension in the regulation of cardiomyocyte cell cycle activity in the postnatal heart. To investigate the role of actomyosin contractility in cardiomyocyte cell cycle arrest, we conditionally activated ROCK2 kinase domain (ROCK2:ER) in the murine postnatal heart. Here, we show that α5/β1 integrin and fibronectin matrix increase in response to actomyosin-mediated tension. Moreover, activation of ROCK2:ER promotes nuclear translocation of Yap, a mechanosensitive transcriptional co-activator, and enhances cardiomyocyte proliferation. Finally, we show that reduction of myocardial α5 integrin rescues the myocardial proliferation phenotype in ROCK2:ER hearts. These data demonstrate that cardiomyocytes respond to increased intracellular tension by altering their intercellular contacts in favor of cell-matrix interactions, leading to Yap nuclear translocation, thus uncovering a function for nonmuscle myosin contractility in promoting cardiomyocyte proliferation in the postnatal heart.
Collapse
Affiliation(s)
- Xiaofei Li
- Cardiovascular Research Center, Lifespan Cardiovascular Institute, Rhode Island Hospital, Department of Medicine, Division of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Callie McLain
- Cardiovascular Research Center, Lifespan Cardiovascular Institute, Rhode Island Hospital, Department of Medicine, Division of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Michael S. Samuel
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Michael F. Olson
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, M5B 2K3 Canada
| | - Glenn L. Radice
- Cardiovascular Research Center, Lifespan Cardiovascular Institute, Rhode Island Hospital, Department of Medicine, Division of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
13
|
Rocha DN, Carvalho ED, Relvas JB, Oliveira MJ, Pêgo AP. Mechanotransduction: Exploring New Therapeutic Avenues in Central Nervous System Pathology. Front Neurosci 2022; 16:861613. [PMID: 35573316 PMCID: PMC9096357 DOI: 10.3389/fnins.2022.861613] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Cells are continuously exposed to physical forces and the central nervous system (CNS) is no exception. Cells dynamically adapt their behavior and remodel the surrounding environment in response to forces. The importance of mechanotransduction in the CNS is illustrated by exploring its role in CNS pathology development and progression. The crosstalk between the biochemical and biophysical components of the extracellular matrix (ECM) are here described, considering the recent explosion of literature demonstrating the powerful influence of biophysical stimuli like density, rigidity and geometry of the ECM on cell behavior. This review aims at integrating mechanical properties into our understanding of the molecular basis of CNS disease. The mechanisms that mediate mechanotransduction events, like integrin, Rho/ROCK and matrix metalloproteinases signaling pathways are revised. Analysis of CNS pathologies in this context has revealed that a wide range of neurological diseases share as hallmarks alterations of the tissue mechanical properties. Therefore, it is our belief that the understanding of CNS mechanotransduction pathways may lead to the development of improved medical devices and diagnostic methods as well as new therapeutic targets and strategies for CNS repair.
Collapse
Affiliation(s)
- Daniela Nogueira Rocha
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Eva Daniela Carvalho
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia (FEUP), Universidade do Porto, Porto, Portugal
| | - João Bettencourt Relvas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Paula Pêgo
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
14
|
Bianchi F, Sommariva M, Cornaghi LB, Denti L, Nava A, Arnaboldi F, Moscheni C, Gagliano N. Mechanical Cues, E-Cadherin Expression and Cell "Sociality" Are Crucial Crossroads in Determining Pancreatic Ductal Adenocarcinoma Cells Behavior. Cells 2022; 11:1318. [PMID: 35455997 PMCID: PMC9028873 DOI: 10.3390/cells11081318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/23/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
E-cadherin, an epithelial-to-mesenchymal transition (EMT) marker, is coupled to actin cytoskeleton and distributes cell forces acting on cells. Since YAP transduces mechanical signals involving actin cytoskeleton, we aimed to investigate the relationship between YAP and mechanical cues in pancreatic ductal adenocarcinoma (PDAC) cell lines, characterized by different EMT-related phenotypes, cultured in 2D monolayers and 3D spheroids. We observed that the YAP/p-YAP ratio was reduced in HPAC and MIA PaCa-2 cell lines and remained unchanged in BxPC-3 cells when cultured in a 3D setting. CTGF and CYR61 gene expression were down-regulated in all PDAC 3D compared to 2D cultures, without any significant effect following actin cytoskeleton inhibition by Cytochalasin B (CyB) treatment. Moreover, LATS1 mRNA, indicating the activation of the Hippo pathway, was not influenced by CyB and differed in all PDAC cell lines having different EMT-related phenotype but a similar pattern of CTGF and CYR61 expression. Although the role of YAP modulation in response to mechanical cues in cancer cells remains to be completely elucidated, our results suggest that cell arrangement and phenotype can determine variable outcomes to mechanical stimuli in PDAC cells. Moreover, it is possible to speculate that YAP and Hippo pathways may act as parallel and not exclusive inputs that, converging at some points, may impact cell behavior.
Collapse
Affiliation(s)
- Francesca Bianchi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (M.S.); (L.B.C.); (A.N.); (F.A.)
- U. O. Laboratorio Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Michele Sommariva
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (M.S.); (L.B.C.); (A.N.); (F.A.)
| | - Laura Brigida Cornaghi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (M.S.); (L.B.C.); (A.N.); (F.A.)
| | - Luca Denti
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Ambra Nava
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (M.S.); (L.B.C.); (A.N.); (F.A.)
| | - Francesca Arnaboldi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (M.S.); (L.B.C.); (A.N.); (F.A.)
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences “L. Sacco”, Università degli Studi di Milano, 20157 Milan, Italy;
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (M.S.); (L.B.C.); (A.N.); (F.A.)
| |
Collapse
|
15
|
Mechanical transmission enables EMT cancer cells to drive epithelial cancer cell migration to guide tumor spheroid disaggregation. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2031-2049. [PMID: 35366152 DOI: 10.1007/s11427-021-2054-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023]
Abstract
Cell phenotype heterogeneity within tumor tissue, especially which due to the emergence of epithelial-mesenchymal transition (EMT) in cancer cells, is associated with cancer invasion and metastasis. However, our understanding of the cellular mechanism(s) underlying the cooperation between EMT cell and epithelial cancer cell migration remains incomplete. Herein, heterotypic tumor spheroids containing both epithelial and EMT cancer cells were generated in vitro. We observed that EMT cells dominated the peripheral region of the self-organized heterotypic tumor spheroid. Furthermore, our results demonstrated that EMT cells could serve as leader cells to improve the collective migration efficiency of epithelial cancer cells and promote dispersion and invasion of the tumor spheroids, which was regulated by the force transition between EMT cells and epithelial cancer cells. Mechanistically, our data further suggest that force transmission is mediated by heterophilic N-cadherin/E-cadherin adhesion complexes between EMT and epithelial cancer cells. Impairment of N-cadherin/E-cadherin adhesion complex formation abrogated the ability of EMT cells to guide epithelial cancer cell migration and blocked the dispersion of tumor spheroids. Together, our data provide new insight into the mechanical interaction between epithelial and EMT cancer cells through heterophilic cadherin adhesion, which enables cooperative tumor cell migration, highlighting the role of EMT cells in tumor invasion.
Collapse
|
16
|
Mierke CT. Viscoelasticity, Like Forces, Plays a Role in Mechanotransduction. Front Cell Dev Biol 2022; 10:789841. [PMID: 35223831 PMCID: PMC8864183 DOI: 10.3389/fcell.2022.789841] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Viscoelasticity and its alteration in time and space has turned out to act as a key element in fundamental biological processes in living systems, such as morphogenesis and motility. Based on experimental and theoretical findings it can be proposed that viscoelasticity of cells, spheroids and tissues seems to be a collective characteristic that demands macromolecular, intracellular component and intercellular interactions. A major challenge is to couple the alterations in the macroscopic structural or material characteristics of cells, spheroids and tissues, such as cell and tissue phase transitions, to the microscopic interferences of their elements. Therefore, the biophysical technologies need to be improved, advanced and connected to classical biological assays. In this review, the viscoelastic nature of cytoskeletal, extracellular and cellular networks is presented and discussed. Viscoelasticity is conceptualized as a major contributor to cell migration and invasion and it is discussed whether it can serve as a biomarker for the cells' migratory capacity in several biological contexts. It can be hypothesized that the statistical mechanics of intra- and extracellular networks may be applied in the future as a powerful tool to explore quantitatively the biomechanical foundation of viscoelasticity over a broad range of time and length scales. Finally, the importance of the cellular viscoelasticity is illustrated in identifying and characterizing multiple disorders, such as cancer, tissue injuries, acute or chronic inflammations or fibrotic diseases.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Zhang C, Zhu H, Ren X, Gao B, Cheng B, Liu S, Sha B, Li Z, Zhang Z, Lv Y, Wang H, Guo H, Lu TJ, Xu F, Genin GM, Lin M. Mechanics-driven nuclear localization of YAP can be reversed by N-cadherin ligation in mesenchymal stem cells. Nat Commun 2021; 12:6229. [PMID: 34711824 PMCID: PMC8553821 DOI: 10.1038/s41467-021-26454-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 10/01/2021] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cells adopt differentiation pathways based upon cumulative effects of mechanosensing. A cell's mechanical microenvironment changes substantially over the course of development, beginning from the early stages in which cells are typically surrounded by other cells and continuing through later stages in which cells are typically surrounded by extracellular matrix. How cells erase the memory of some of these mechanical microenvironments while locking in memory of others is unknown. Here, we develop a material and culture system for modifying and measuring the degree to which cells retain cumulative effects of mechanosensing. Using this system, we discover that effects of the RGD adhesive motif of fibronectin (representative of extracellular matrix), known to impart what is often termed "mechanical memory" in mesenchymal stem cells via nuclear YAP localization, are erased by the HAVDI adhesive motif of the N-cadherin (representative of cell-cell contacts). These effects can be explained by a motor clutch model that relates cellular traction force, nuclear deformation, and resulting nuclear YAP re-localization. Results demonstrate that controlled storage and removal of proteins associated with mechanical memory in mesenchymal stem cells is possible through defined and programmable material systems.
Collapse
Affiliation(s)
- Cheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Xinru Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Bin Gao
- Department of Endocrinology, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, People's Republic of China
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Shaobao Liu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, People's Republic of China
| | - Baoyong Sha
- School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, People's Republic of China
| | - Zhaoqing Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Zheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, People's Republic of China
| | - Haohua Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, People's Republic of China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, People's Republic of China
- MOE Key Laboratory of Multifunctional Materials and Structures, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, 63130, MO, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, 63130, MO, USA
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| |
Collapse
|
18
|
Cole A, Buckler S, Marcucci J, Artemenko Y. Differential Roles of Actin Crosslinking Proteins Filamin and α-Actinin in Shear Flow-Induced Migration of Dictyostelium discoideum. Front Cell Dev Biol 2021; 9:743011. [PMID: 34485315 PMCID: PMC8415421 DOI: 10.3389/fcell.2021.743011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 07/28/2021] [Indexed: 01/28/2023] Open
Abstract
Shear flow-induced migration is an important physiological phenomenon experienced by multiple cell types, including leukocytes and cancer cells. However, molecular mechanisms by which cells sense and directionally migrate in response to mechanical perturbation are not well understood. Dictyostelium discoideum social amoeba, a well-established model for studying amoeboid-type migration, also exhibits directional motility when exposed to shear flow, and this behavior is preceded by rapid and transient activation of the same signal transduction network that is activated by chemoattractants. The initial response, which can also be observed following brief 2 s stimulation with shear flow, requires an intact actin cytoskeleton; however, what aspect of the cytoskeletal network is responsible for sensing and/or transmitting the signal is unclear. We investigated the role of actin crosslinkers filamin and α-actinin by analyzing initial shear flow-stimulated responses in cells with or without these proteins. Both filamin and α-actinin showed rapid and transient relocalization from the cytosol to the cortex following shear flow stimulation. Using spatiotemporal analysis of Ras GTPase activation as a readout of signal transduction network activity, we demonstrated that lack of α-actinin did not reduce, and, in fact, slightly improved the response to acute mechanical stimulation compared to cells expressing α-actinin. In contrast, shear flow-induced Ras activation was significantly more robust in filamin-null cells rescued with filamin compared to cells expressing empty vector. Reduced responsiveness appeared to be specific to mechanical stimuli and was not due to a change in the basal activity since response to global stimulation with a chemoattractant and random migration was comparable between cells with or without filamin. Finally, while filamin-null cells rescued with filamin efficiently migrated upstream when presented with continuous flow, cells lacking filamin were defective in directional migration. Overall, our study suggests that filamin, but not α-actinin, is involved in sensing and/or transmitting mechanical stimuli that drive directed migration; however, other components of the actin cytoskeleton likely also contribute to the initial response since filamin-null cells were still able to activate the signal transduction network. These findings could have implications for our fundamental understanding of shear flow-induced migration of leukocytes, cancer cells and other amoeboid-type cells.
Collapse
Affiliation(s)
- Aaron Cole
- Department of Biological Sciences, State University of New York Oswego, Oswego, NY, United States
| | - Sarah Buckler
- Department of Biological Sciences, State University of New York Oswego, Oswego, NY, United States
| | - Jack Marcucci
- Department of Biological Sciences, State University of New York Oswego, Oswego, NY, United States
| | - Yulia Artemenko
- Department of Biological Sciences, State University of New York Oswego, Oswego, NY, United States
| |
Collapse
|
19
|
Hang X, He S, Dong Z, Minnick G, Rosenbohm J, Chen Z, Yang R, Chang L. Nanosensors for single cell mechanical interrogation. Biosens Bioelectron 2021; 179:113086. [DOI: 10.1016/j.bios.2021.113086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 02/08/2023]
|
20
|
Lian M, Sun B, Han Y, Yu B, Xin W, Xu R, Ni B, Jiang W, Hao Y, Zhang X, Shen Y, Qiao Z, Dai K. A low-temperature-printed hierarchical porous sponge-like scaffold that promotes cell-material interaction and modulates paracrine activity of MSCs for vascularized bone regeneration. Biomaterials 2021; 274:120841. [PMID: 33984633 DOI: 10.1016/j.biomaterials.2021.120841] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/31/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) secrete paracrine trophic factors that are beneficial for tissue regeneration. In this study, a sponge-like scaffold with hierarchical and interconnected pores was developed using low-temperature deposition modeling (LDM) printing. Its effects on the cellular behavior, especially on the paracrine secretion patterns of MSCs, were comprehensively investigated. We found that compared with the scaffolds printed via the fused deposition modeling (FDM) technique, the LDM-printed sponges enhanced the adhesion, retention, survival, and ingrowth of MSCs and promoted cell-material interactions. Moreover, the paracrine functions of the cultured MSCs on the LDM-printed sponges were improved, with significant secretion of upregulated immunomodulatory, angiogenic, and osteogenic factors. MSCs on the LDM-printed sponges exert beneficial paracrine effects on multiple regenerative processes, including macrophage polarization, tube formation, and osteogenesis, verifying the enhanced immunomodulatory, angiogenic, and osteogenic potential. Further protein function assays indicated that focal adhesion kinase (FAK), downstream AKT, and yes-associated-protein (YAP) signaling might participate in the required mechanotransductive pathways, through which the hierarchical porous structures stimulated the paracrine effects of MSCs. In a rat distal femoral defect model, the MSC-laden LDM-printed sponges significantly promoted vascularized bone regeneration. The results of the present study demonstrate that the hierarchical porous biomimetic sponges prepared via LDM printing have potential applications in tissue engineering based on their cell-material interaction promotion and MSC paracrine function modulation effects. Furthermore, our findings suggest that the optimization of biomaterial properties to direct the paracrine signaling of MSCs would enhance tissue regeneration.
Collapse
Affiliation(s)
- Meifei Lian
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Binbin Sun
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yu Han
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bin Yu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weiwei Xin
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China
| | - Ruida Xu
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China
| | - Bing Ni
- School of Life Science, East China Normal University, Shanghai, 200241, China
| | - Wenbo Jiang
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yongqiang Hao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiuyin Zhang
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi Shen
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China.
| | - Zhiguang Qiao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China.
| | - Kerong Dai
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
21
|
Ectoderm to mesoderm transition by down-regulation of actomyosin contractility. PLoS Biol 2021; 19:e3001060. [PMID: 33406067 PMCID: PMC7815211 DOI: 10.1371/journal.pbio.3001060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 01/19/2021] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
Collective migration of cohesive tissues is a fundamental process in morphogenesis and is particularly well illustrated during gastrulation by the rapid and massive internalization of the mesoderm, which contrasts with the much more modest movements of the ectoderm. In the Xenopus embryo, the differences in morphogenetic capabilities of ectoderm and mesoderm can be connected to the intrinsic motility of individual cells, very low for ectoderm, high for mesoderm. Surprisingly, we find that these seemingly deep differences can be accounted for simply by differences in Rho-kinases (Rock)-dependent actomyosin contractility. We show that Rock inhibition is sufficient to rapidly unleash motility in the ectoderm and confer it with mesoderm-like properties. In the mesoderm, this motility is dependent on two negative regulators of RhoA, the small GTPase Rnd1 and the RhoGAP Shirin/Dlc2/ArhGAP37. Both are absolutely essential for gastrulation. At the cellular and tissue level, the two regulators show overlapping yet distinct functions. They both contribute to decrease cortical tension and confer motility, but Shirin tends to increase tissue fluidity and stimulate dispersion, while Rnd1 tends to favor more compact collective migration. Thus, each is able to contribute to a specific property of the migratory behavior of the mesoderm. We propose that the "ectoderm to mesoderm transition" is a prototypic case of collective migration driven by a down-regulation of cellular tension, without the need for the complex changes traditionally associated with the epithelial-to-mesenchymal transition.
Collapse
|
22
|
López-Gay JM, Nunley H, Spencer M, di Pietro F, Guirao B, Bosveld F, Markova O, Gaugue I, Pelletier S, Lubensky DK, Bellaïche Y. Apical stress fibers enable a scaling between cell mechanical response and area in epithelial tissue. Science 2020; 370:370/6514/eabb2169. [PMID: 33060329 DOI: 10.1126/science.abb2169] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022]
Abstract
Biological systems tailor their properties and behavior to their size throughout development and in numerous aspects of physiology. However, such size scaling remains poorly understood as it applies to cell mechanics and mechanosensing. By examining how the Drosophila pupal dorsal thorax epithelium responds to morphogenetic forces, we found that the number of apical stress fibers (aSFs) anchored to adherens junctions scales with cell apical area to limit larger cell elongation under mechanical stress. aSFs cluster Hippo pathway components, thereby scaling Hippo signaling and proliferation with area. This scaling is promoted by tricellular junctions mediating an increase in aSF nucleation rate and lifetime in larger cells. Development, homeostasis, and repair entail epithelial cell size changes driven by mechanical forces; our work highlights how, in turn, mechanosensitivity scales with cell size.
Collapse
Affiliation(s)
- Jesús M López-Gay
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Hayden Nunley
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Meryl Spencer
- Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Florencia di Pietro
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Boris Guirao
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Floris Bosveld
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Olga Markova
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Isabelle Gaugue
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - Stéphane Pelletier
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France.,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| | - David K Lubensky
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA. .,Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yohanns Bellaïche
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, F-75248 Paris Cedex 05, France. .,Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, F-75005 Paris, France
| |
Collapse
|
23
|
Khalil AA, Ilina O, Vasaturo A, Venhuizen JH, Vullings M, Venhuizen V, Bilos A, Figdor CG, Span PN, Friedl P. Collective invasion induced by an autocrine purinergic loop through connexin-43 hemichannels. J Cell Biol 2020; 219:e201911120. [PMID: 32777015 PMCID: PMC7659730 DOI: 10.1083/jcb.201911120] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 05/23/2020] [Accepted: 06/30/2020] [Indexed: 02/08/2023] Open
Abstract
Progression of epithelial cancers predominantly proceeds by collective invasion of cell groups with coordinated cell-cell junctions and multicellular cytoskeletal activity. Collectively invading breast cancer cells express the gap junction protein connexin-43 (Cx43), yet whether Cx43 regulates collective invasion remains unclear. We here show that Cx43 mediates gap-junctional coupling between collectively invading breast cancer cells and, via hemichannels, adenosine nucleotide/nucleoside release into the extracellular space. Using molecular interference and rescue strategies, we identify that Cx43 hemichannel function, but not intercellular communication, induces leader cell activity and collective migration through the engagement of the adenosine receptor 1 (ADORA1) and AKT signaling. Accordingly, pharmacological inhibition of ADORA1 or AKT signaling caused leader cell collapse and halted collective invasion. ADORA1 inhibition further reduced local invasion of orthotopic mammary tumors in vivo, and joint up-regulation of Cx43 and ADORA1 in breast cancer patients correlated with decreased relapse-free survival. This identifies autocrine purinergic signaling, through Cx43 hemichannels, as a critical pathway in leader cell function and collective invasion.
Collapse
Affiliation(s)
- Antoine A. Khalil
- Department of Dermatology and Graduate School of Life Science, University of Wuerzburg, Wuerzburg, Germany
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Olga Ilina
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Angela Vasaturo
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jan-Hendrik Venhuizen
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Manon Vullings
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Victor Venhuizen
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ab Bilos
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul N. Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Friedl
- Department of Dermatology and Graduate School of Life Science, University of Wuerzburg, Wuerzburg, Germany
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- David H. Koch Center for Genitourinary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
- Cancer Genomics Center, Utrecht, Netherlands
| |
Collapse
|
24
|
Zuidema A, Wang W, Sonnenberg A. Crosstalk between Cell Adhesion Complexes in Regulation of Mechanotransduction. Bioessays 2020; 42:e2000119. [PMID: 32830356 DOI: 10.1002/bies.202000119] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/27/2020] [Indexed: 01/03/2023]
Abstract
Physical forces regulate numerous biological processes during development, physiology, and pathology. Forces between the external environment and intracellular actin cytoskeleton are primarily transmitted through integrin-containing focal adhesions and cadherin-containing adherens junctions. Crosstalk between these complexes is well established and modulates the mechanical landscape of the cell. However, integrins and cadherins constitute large families of adhesion receptors and form multiple complexes by interacting with different ligands, adaptor proteins, and cytoskeletal filaments. Recent findings indicate that integrin-containing hemidesmosomes oppose force transduction and traction force generation by focal adhesions. The cytolinker plectin mediates this crosstalk by coupling intermediate filaments to the actin cytoskeleton. Similarly, cadherins in desmosomes might modulate force generation by adherens junctions. Moreover, mechanotransduction can be influenced by podosomes, clathrin lattices, and tetraspanin-enriched microdomains. This review discusses mechanotransduction by multiple integrin- and cadherin-based cell adhesion complexes, which together with the associated cytoskeleton form an integrated network that allows cells to sense, process, and respond to their physical environment.
Collapse
Affiliation(s)
- Alba Zuidema
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Wei Wang
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Arnoud Sonnenberg
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| |
Collapse
|
25
|
Maddala R, Rao PV. Global phosphotyrosinylated protein profile of cell-matrix adhesion complexes of trabecular meshwork cells. Am J Physiol Cell Physiol 2020; 319:C288-C299. [PMID: 32432933 PMCID: PMC7500213 DOI: 10.1152/ajpcell.00537.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/27/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022]
Abstract
Dysregulation of the mechanical properties and cell adhesive interactions of trabecular meshwork (TM) are known to impair aqueous humor drainage and elevate intraocular pressure in glaucoma patients. The identity of regulatory mechanisms underlying TM mechanotransduction, however, remains elusive. Here we analyzed the phosphotyrosine proteome of human TM cell-extracellular matrix (ECM) adhesion complexes, which play a key role in sensing and transducing extracellular chemical and mechanical cues into intracellular activities, using a two-level affinity pull-down (phosphotyrosine antibody and titanium dioxide beads) method and mass spectrometry. This analysis identified ~1,000 tyrosine-phosphorylated proteins of TM cell-ECM adhesion complexes. Many consensus adhesome proteins were found to be tyrosine phosphorylated. Interestingly, several of the phosphotyrosinylated proteins found in TM cell-ECM adhesion complexes are known to be required for podocyte glomerular filtration, indicating the existence of molecular parallels that are likely relevant to the shared fluid barrier and filtration functions of the two mechanosensitive cell types.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
26
|
The intercalated disc: a mechanosensing signalling node in cardiomyopathy. Biophys Rev 2020; 12:931-946. [PMID: 32661904 PMCID: PMC7429531 DOI: 10.1007/s12551-020-00737-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/08/2020] [Indexed: 02/08/2023] Open
Abstract
Cardiomyocytes, the cells generating contractile force in the heart, are connected to each other through a highly specialised structure, the intercalated disc (ID), which ensures force transmission and transduction between neighbouring cells and allows the myocardium to function in synchrony. In addition, cardiomyocytes possess an intrinsic ability to sense mechanical changes and to regulate their own contractile output accordingly. To achieve this, some of the components responsible for force transmission have evolved to sense changes in tension and to trigger a biochemical response that results in molecular and cellular changes in cardiomyocytes. This becomes of particular importance in cardiomyopathies, where the heart is exposed to increased mechanical load and needs to adapt to sustain its contractile function. In this review, we will discuss key mechanosensing elements present at the intercalated disc and provide an overview of the signalling molecules involved in mediating the responses to changes in mechanical force.
Collapse
|
27
|
Serna-Márquez N, Rodríguez-Hernández A, Ayala-Reyes M, Martínez-Hernández LO, Peña-Rico MÁ, Carretero-Ortega J, Hautefeuille M, Vázquez-Victorio G. Fibrillar Collagen Type I Participates in the Survival and Aggregation of Primary Hepatocytes Cultured on Soft Hydrogels. Biomimetics (Basel) 2020; 5:E30. [PMID: 32630500 PMCID: PMC7345357 DOI: 10.3390/biomimetics5020030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Liver is an essential organ that carries out multiple functions such as glycogen storage, the synthesis of plasma proteins, and the detoxification of xenobiotics. Hepatocytes are the parenchyma that sustain almost all the functions supported by this organ. Hepatocytes and non-parenchymal cells respond to the mechanical alterations that occur in the extracellular matrix (ECM) caused by organogenesis and regenerating processes. Rearrangements of the ECM modify the composition and mechanical properties that result in specific dedifferentiation programs inside the hepatic cells. Quiescent hepatocytes are embedded in the soft ECM, which contains an important concentration of fibrillar collagens in combination with a basement membrane-associated matrix (BM). This work aims to evaluate the role of fibrillar collagens and BM on actin cytoskeleton organization and the function of rat primary hepatocytes cultured on soft elastic polyacrylamide hydrogels (PAA HGs). We used rat tail collagen type I and Matrigel® as references of fibrillar collagens and BM respectively and mixed different percentages of collagen type I in combination with BM. We also used peptides obtained from decellularized liver matrices (dECM). Remarkably, hepatocytes showed a poor adhesion in the absence of collagen on soft PAA HGs. We demonstrated that collagen type I inhibited apoptosis and activated extracellular signal-regulated kinases 1/2 (ERK1/2) in primary hepatocytes cultured on soft hydrogels. Epidermal growth factor (EGF) was not able to rescue cell viability in conjugated BM but affected cell aggregation in soft PAA HGs conjugated with combinations of different proportions of collagen and BM. Interestingly, actin cytoskeleton was localized and preserved close to plasma membrane (cortical actin) and proximal to intercellular ducts (canaliculi-like structures) in soft conditions; however, albumin protein expression was not preserved, even though primary hepatocytes did not remodel their actin cytoskeleton significantly in soft conditions. This investigation highlights the important role of fibrillar collagens on soft hydrogels for the maintenance of survival and aggregation of the hepatocytes. Data suggest evaluating the conditions that allow the establishment of optimal biomimetic environments for physiology and cell biology studies, where the phenotype of primary cells may be preserved for longer periods of time.
Collapse
Affiliation(s)
- Nathalia Serna-Márquez
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Adriana Rodríguez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Marisol Ayala-Reyes
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Lorena Omega Martínez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Miguel Ángel Peña-Rico
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Jorge Carretero-Ortega
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Mathieu Hautefeuille
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| | - Genaro Vázquez-Victorio
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| |
Collapse
|
28
|
Griffith CM, Huang SA, Cho C, Khare TM, Rich M, Lee GH, Ligler FS, Diekman BO, Polacheck WJ. Microfluidics for the study of mechanotransduction. JOURNAL OF PHYSICS D: APPLIED PHYSICS 2020; 53:224004. [PMID: 33840837 PMCID: PMC8034607 DOI: 10.1088/1361-6463/ab78d4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Mechanical forces regulate a diverse set of biological processes at cellular, tissue, and organismal length scales. Investigating the cellular and molecular mechanisms that underlie the conversion of mechanical forces to biological responses is challenged by limitations of traditional animal models and in vitro cell culture, including poor control over applied force and highly artificial cell culture environments. Recent advances in fabrication methods and material processing have enabled the development of microfluidic platforms that provide precise control over the mechanical microenvironment of cultured cells. These devices and systems have proven to be powerful for uncovering and defining mechanisms of mechanotransduction. In this review, we first give an overview of the main mechanotransduction pathways that function at sites of cell adhesion, many of which have been investigated with microfluidics. We then discuss how distinct microfluidic fabrication methods can be harnessed to gain biological insight, with description of both monolithic and replica molding approaches. Finally, we present examples of how microfluidics can be used to apply both solid forces (substrate mechanics, strain, and compression) and fluid forces (luminal, interstitial) to cells. Throughout the review, we emphasize the advantages and disadvantages of different fabrication methods and applications of force in order to provide perspective to investigators looking to apply forces to cells in their own research.
Collapse
Affiliation(s)
- Christian M Griffith
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Stephanie A Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Crescentia Cho
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Tanmay M Khare
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC
| | - Matthew Rich
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Gi-Hun Lee
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Frances S Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- McAllister Heart Institute, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
- Cancer Cell Biology Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| |
Collapse
|
29
|
Galigniana NM, Charó NL, Uranga R, Cabanillas AM, Piwien-Pilipuk G. Oxidative stress induces transcription of telomeric repeat-containing RNA (TERRA) by engaging PKA signaling and cytoskeleton dynamics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118643. [DOI: 10.1016/j.bbamcr.2020.118643] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/27/2019] [Accepted: 01/02/2020] [Indexed: 12/11/2022]
|
30
|
Manet S, Vannier D, Bouin AP, Lisowska J, Albiges-Rizo C, Faurobert E. Immunofluorescence of Cell-Cell and Cell-Extracellular Matrix Adhesive Defects in In Vitro Endothelial CCM Model: Juxtacrine Role of Mutant Extracellular Matrix on Wild-Type Endothelial Cells. Methods Mol Biol 2020; 2152:401-416. [PMID: 32524568 DOI: 10.1007/978-1-0716-0640-7_29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cells lining cerebral cavernous malformations (CCM) present strong adhesive and mechanical defects. Increased cell contractility is a driver to the onset and the expansion of the CCM lesions. 2D in vitro endothelial models have been developed from either endothelial cells isolated from ccm1-3 knock-out mice or CCM1-3-silenced primary endothelial cells. These in vitro models faithfully recapitulate the adhesive and contractile defects of the CCM-deficient endothelial cells such as increased cell-extracellular matrix (ECM) adhesion through β1 integrin-anchored actin stress fibers, abnormal remodeling of the ECM, and destabilized VE-cadherin-dependent cell-cell junctions. Using such 2D in vitro CCM models, we have shown that the ECM remodeled by CCM-depleted endothelial cells can propagate CCM-like adhesive defects to wild-type endothelial cells, a process potentially pertinent to CCM lesion expansion. Here, we detail methods for studying the morphology of focal adhesions, actomyosin cytoskeleton, and VE-cadherin-dependent Adherens junctions by immunofluorescence and morphometric analyses. Moreover, we detail the protocols to produce and purify remodeled ECM and to test its effect on endothelial cell adhesion.
Collapse
Affiliation(s)
- Sandra Manet
- Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Daphné Vannier
- Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Anne-Pascale Bouin
- Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Justyna Lisowska
- Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Corinne Albiges-Rizo
- Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Eva Faurobert
- Institute for Advanced Biosciences, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France.
| |
Collapse
|
31
|
Gou X, E JC, Yang H, Sun D. Combined Single-Cell Manipulation and Chemomechanical Modeling to Probe Cell Migration Mechanism During Cell-to-Cell Interaction. IEEE Trans Biomed Eng 2019; 67:1474-1482. [PMID: 31484106 DOI: 10.1109/tbme.2019.2938569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Spatial presentations of chemical and mechanical information are key parameters for cell migration. However, previous theoretical and experimental studies focus on probing the mechanisms caused by a single type of stimulus, while ignoring the synergetic effects, especially for single cell migration during cell-to-cell interaction. Here we develop a chemomechanical model to assess the biochemical and biophysical modulators of single cell migration during cell-to-cell interaction. This model considers the stimulation of chemoattractant concentration gradient, influence of dynamic adhesion strength and relative motion between cells. The model is validated with single cell manipulation of leukemia cancer cell on stromal cell layer using optical tweezers. Both the modeling and experimental results demonstrate that cell migration velocity caused by chemotaxis can be biased by dynamic adhesion force, which is related to the retrograde flow of stromal cell layer. Besides, the biophysical modulators can influence the effect of drug treatment for specific signaling pathway. Our work provides a quantitative description of single cell migration in a complex environment that is close to realistic in vivo situation and is useful for further exploration of cell signaling pathway during cell-to-cell interactions for investigation of potential therapeutic strategy.
Collapse
|
32
|
Xie Y, Zhang L, Xiong Q, Gao Y, Ge W, Tang P. Bench-to-bedside strategies for osteoporotic fracture: From osteoimmunology to mechanosensation. Bone Res 2019; 7:25. [PMID: 31646015 PMCID: PMC6804735 DOI: 10.1038/s41413-019-0066-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 12/16/2022] Open
Abstract
Osteoporosis is characterized by a decrease in bone mass and strength, rendering people prone to osteoporotic fractures caused by low-energy forces. The primary treatment strategy for osteoporotic fractures is surgery; however, the compromised and comminuted bones in osteoporotic fracture sites are not conducive to optimum reduction and rigid fixation. In addition, these patients always exhibit accompanying aging-related disorders, including high inflammatory status, decreased mechanical loading and abnormal skeletal metabolism, which are disadvantages for fracture healing around sites that have undergone orthopedic procedures. Since the incidence of osteoporosis is expected to increase worldwide, orthopedic surgeons should pay more attention to comprehensive strategies for improving the poor prognosis of osteoporotic fractures. Herein, we highlight the molecular basis of osteoimmunology and bone mechanosensation in different healing phases of elderly osteoporotic fractures, guiding perioperative management to alleviate the unfavorable effects of insufficient mechanical loading, high inflammatory levels and pathogen infection. The well-informed pharmacologic and surgical intervention, including treatment with anti-inflammatory drugs and sufficient application of antibiotics, as well as bench-to-bedside strategies for bone augmentation and hardware selection, should be made according to a comprehensive understanding of bone biomechanical properties in addition to the remodeling status of osteoporotic bones, which is necessary for creating proper biological and mechanical environments for bone union and remodeling. Multidisciplinary collaboration will facilitate the improvement of overall osteoporotic care and reduction of secondary fracture incidence.
Collapse
Affiliation(s)
- Yong Xie
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Qi Xiong
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Yanpan Gao
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
33
|
Chen DY, Sun NH, Lu YP, Hong LJ, Cui TT, Wang CK, Chen XH, Wang SS, Feng LL, Shi WX, Fukunaga K, Chen Z, Lu YM, Han F. GPR124 facilitates pericyte polarization and migration by regulating the formation of filopodia during ischemic injury. Theranostics 2019; 9:5937-5955. [PMID: 31534530 PMCID: PMC6735362 DOI: 10.7150/thno.34168] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/28/2019] [Indexed: 12/31/2022] Open
Abstract
Prolonged occlusion of multiple microvessels causes microvascular injury. G protein-coupled receptor 124 (GPR124) has been reported to be required for maintaining central nervous system (CNS) angiogenesis and blood-brain barrier integrity. However, the molecular mechanisms by which GPR124 regulates pericytes during ischemia have remained elusive. Methods: A microsphere embolism-induced ischemia model was used to evaluate the expression of GPR124 following microsphere embolism. Immunocytochemistry and stochastic optical reconstruction microscopy imaging were used to assess the expression and distribution of GPR124 in human brain vascular pericytes (HBVPs) and after the treatment with 3-morpholino-sydnonimine (SIN-1) or oxygen-glucose deprivation (OGD). The effect of GPR124 knockdown or overexpression on HBVP migration was analyzed in vitro using wound healing assays and a microfluidic device. GPR124 loss-of-function studies were performed in HBVPs and HEK293 cells using CRISPR-Cas9-mediated gene deletion. Time-lapse imaging was used to assess dynamic changes in the formation of filopodia in an individual cell. Finally, to explore the functional domains required for GPR124 activity, deletion mutants were constructed for each of the N-terminal domains. Results: GPR124 expression was increased in pericytes following microsphere embolism. Morphological analysis showed localization of GPR124 to focal adhesions where GPR124 bound directly to the actin binding protein vinculin and upregulated Cdc42. SIN-1 or OGD treatment redistributed GPR124 to the leading edges of HBVPs where GPR124 signaling was required for pericyte filopodia formation and directional migration. Partial deletion of GPR124 domains decreased SIN-1-induced filopodia formation and cell migration. Conclusion: Taken together, our results provide the first evidence for a role of GPR124 in pericyte migration under ischemic conditions and suggest that GPR124 was essential for Cdc42 activation and filopodia formation.
Collapse
|
34
|
Abstract
Mechanical forces drive the remodeling of tissues during morphogenesis. This relies on the transmission of forces between cells by cadherin-based adherens junctions, which couple the force-generating actomyosin cytoskeletons of neighboring cells. Moreover, components of cadherin adhesions adopt force-dependent conformations that induce changes in the composition of adherens junctions, enabling transduction of mechanical forces into an intracellular response. Cadherin mechanotransduction can mediate reinforcement of cell–cell adhesions to withstand forces but also induce biochemical signaling to regulate cell behavior or direct remodeling of cell–cell adhesions to enable cell rearrangements. By transmission and transduction of mechanical forces, cadherin adhesions coordinate cellular behaviors underlying morphogenetic processes of collective cell migration, cell division, and cell intercalation. Here, we review recent advances in our understanding of this central role of cadherin adhesions in force-dependent regulation of morphogenesis.
Collapse
Affiliation(s)
- Willem-Jan Pannekoek
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn Gloerich
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
35
|
Uncoupling Traditional Functionalities of Metastasis: The Parting of Ways with Real-Time Assays. J Clin Med 2019; 8:jcm8070941. [PMID: 31261795 PMCID: PMC6678138 DOI: 10.3390/jcm8070941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
The experimental evaluation of metastasis overly focuses on the gain of migratory and invasive properties, while disregarding the contributions of cellular plasticity, extra-cellular matrix heterogeneity, niche interactions, and tissue architecture. Traditional cell-based assays often restrict the inclusion of these processes and warrant the implementation of approaches that provide an enhanced spatiotemporal resolution of the metastatic cascade. Time lapse imaging represents such an underutilized approach in cancer biology, especially in the context of disease progression. The inclusion of time lapse microscopy and microfluidic devices in routine assays has recently discerned several nuances of the metastatic cascade. Our review emphasizes that a complete comprehension of metastasis in view of evolving ideologies necessitates (i) the use of appropriate, context-specific assays and understanding their inherent limitations; (ii) cautious derivation of inferences to avoid erroneous/overestimated clinical extrapolations; (iii) corroboration between multiple assay outputs to gauge metastatic potential; and (iv) the development of protocols with improved in situ implications. We further believe that the adoption of improved quantitative approaches in these assays can generate predictive algorithms that may expedite therapeutic strategies targeting metastasis via the development of disease relevant model systems. Such approaches could potentiate the restructuring of the cancer metastasis paradigm through an emphasis on the development of next-generation real-time assays.
Collapse
|
36
|
Liu X, Hou W, He L, Han F, Lu M, Lu X, Duan K, Guo T, Weng J. AMOT130/YAP pathway in topography-induced BMSC osteoblastic differentiation. Colloids Surf B Biointerfaces 2019; 182:110332. [PMID: 31325776 DOI: 10.1016/j.colsurfb.2019.06.061] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/07/2019] [Accepted: 06/26/2019] [Indexed: 02/04/2023]
Abstract
Micro/nano-topography (MNT) is an important variable affecting osseointegration of bone biomaterials, but the underlying mechanisms are not fully understood. We probed the role of a AMOT130/YAP pathway in osteoblastic differentiation of bone marrow mesenchymal stems cultured on titanium (Ti) carrying MNTs. Ti surfaces with two well-defined MNTs (TiO2 nanotubes of different diameters and wall thicknesses) were prepared by anodization. Rat BMSCs were cultured on flat Ti and Ti surfaces carrying MNTs, and cell behaviors (i.e., morphology, F-actin development, osteoblastic differentiation, YAP localization) were studied. Ti surfaces carrying MNTs increased F-actin formation, osteoblastic gene expression, and protein AMOT130 production in BMSCs (all vs. flat Ti), and the surface carrying larger nantubes was more effective, confirming osteoblastic differentiation induced by MNTs. Elevation of the AMOT130 level (by inhibiting its degradation) increased the osteoblastic gene expression, F-actin formation, and nuclear localization of YAP. These show that, AMOT130/YAP is an important pathway mediating the translation of MNT signals to BMSC osteoblastic commitment, likely via the cascade: AMOT130 promotion of F-actin formation, increased YAP nuclear import, and activation of osteoblastic gene expression.
Collapse
Affiliation(s)
- Xuan Liu
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenqing Hou
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Lei He
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Fangping Han
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Mengjie Lu
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Orthopaedic Engineering, Department of Bone and Joint Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiaobo Lu
- Laboratory of Orthopaedic Engineering, Department of Bone and Joint Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ke Duan
- Laboratory of Orthopaedic Engineering, Department of Bone and Joint Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Tailin Guo
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Jie Weng
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
37
|
Hennigan RF, Fletcher JS, Guard S, Ratner N. Proximity biotinylation identifies a set of conformation-specific interactions between Merlin and cell junction proteins. Sci Signal 2019; 12:12/578/eaau8749. [PMID: 31015291 DOI: 10.1126/scisignal.aau8749] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurofibromatosis type 2 is an inherited, neoplastic disease associated with schwannomas, meningiomas, and ependymomas and that is caused by inactivation of the tumor suppressor gene NF2 The NF2 gene product, Merlin, has no intrinsic catalytic activity; its tumor suppressor function is mediated through the proteins with which it interacts. We used proximity biotinylation followed by mass spectrometry and direct binding assays to identify proteins that associated with wild-type and various mutant forms of Merlin in immortalized Schwann cells. We defined a set of 52 proteins in close proximity to wild-type Merlin. Most of the Merlin-proximal proteins were components of cell junctional signaling complexes, suggesting that additional potential interaction partners may exist in adherens junctions, tight junctions, and focal adhesions. With mutant forms of Merlin that cannot bind to phosphatidylinositol 4,5-bisphosphate (PIP2) or that constitutively adopt a closed conformation, we confirmed a critical role for PIP2 binding in Merlin function and identified a large cohort of proteins that specifically interacted with Merlin in the closed conformation. Among these proteins, we identified a previously unreported Merlin-binding protein, apoptosis-stimulated p53 protein 2 (ASPP2, also called Tp53bp2), that bound to closed-conformation Merlin predominately through the FERM domain. Our results demonstrate that Merlin is a component of cell junctional mechanosensing complexes and defines a specific set of proteins through which it acts.
Collapse
Affiliation(s)
- Robert F Hennigan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA.
| | - Jonathan S Fletcher
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Steven Guard
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| |
Collapse
|
38
|
Bijonowski BM, Daraiseh SI, Yuan X, Ma T. Size-Dependent Cortical Compaction Induces Metabolic Adaptation in Mesenchymal Stem Cell Aggregates. Tissue Eng Part A 2019; 25:575-587. [PMID: 30187829 PMCID: PMC6482905 DOI: 10.1089/ten.tea.2018.0155] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/31/2018] [Indexed: 01/01/2023] Open
Abstract
IMPACT STATEMENT This study reveals that multicellular aggregation induces metabolic reprogramming via mechanical compaction in lieu of formation of a hypoxic core. Utilizing biomechanical knowledge gained from planar culture, we set forth a novel three-dimensional (3D) model of size-dependent cortical compaction and demonstrated its role in metabolic reconfiguration. Ultimately, this study establishes mechanical compaction and its spatial gradients as key regulatory factors and design parameters in the development of 3D human adipose-derived mesenchymal stem cell aggregates.
Collapse
Affiliation(s)
- Brent M. Bijonowski
- Dept. of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Susan I. Daraiseh
- Dept. of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| | - Xuegang Yuan
- Dept. of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Teng Ma
- Dept. of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| |
Collapse
|
39
|
From cell shape to cell fate via the cytoskeleton - Insights from the epidermis. Exp Cell Res 2019; 378:232-237. [PMID: 30872138 DOI: 10.1016/j.yexcr.2019.03.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/02/2019] [Accepted: 03/08/2019] [Indexed: 12/31/2022]
Abstract
Animal cells exhibit a wide range of shapes that reflect their diverse functions. Cell shape is determined by a balance between internal and external forces and therefore involves the cytoskeleton and its associated adhesion structures. Cell shape dynamics during development and homeostasis are tightly regulated and closely coordinated with cell fate determination. Defects in cell shape are a hallmark of many pathological conditions including cancer and skin diseases. This review highlights the links between cell shape and cell fate in the epidermis, which have been studied for over 40 years both in vitro and in vivo. Briefly discussing seminal experiments showing the strong coupling between keratinocyte cell shape and their fate we primarily focus on recent studies uncovering novel cellular and molecular mechanisms linking epidermal cell shape with cell growth, differentiation, asymmetric division, and delamination.
Collapse
|
40
|
Humphries JD, Chastney MR, Askari JA, Humphries MJ. Signal transduction via integrin adhesion complexes. Curr Opin Cell Biol 2019; 56:14-21. [PMID: 30195153 DOI: 10.1016/j.ceb.2018.08.004] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/19/2022]
Abstract
Integrin adhesion complexes (IACs) have evolved over millions of years to integrate metazoan cells physically with their microenvironment. It is presumed that the simultaneous interaction of thousands of integrin receptors to binding sites in anisotropic extracellular matrix (ECM) networks enables cells to assemble a topological description of the chemical and mechanical properties of their surroundings. This information is then converted into intracellular signals that influence cell positioning, differentiation and growth, but may also influence other fundamental processes, such as protein synthesis and energy regulation. In this way, changes in the microenvironment can influence all aspects of cell phenotype. Current concepts envisage cell fate decisions being controlled by the integrated signalling output of myriad receptor clusters, but the mechanisms are not understood. Analyses of the adhesome, the complement of proteins attracted to the vicinity of IACs, are now providing insights into some of the primordial links connecting these processes. This article reviews recent advances in our understanding of the composition of IACs, the mechanisms used to transduce signals through these junctions, and the links between IACs and cell phenotype.
Collapse
Affiliation(s)
- Jonathan D Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Megan R Chastney
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Janet A Askari
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
41
|
Kanoldt V, Fischer L, Grashoff C. Unforgettable force – crosstalk and memory of mechanosensitive structures. Biol Chem 2018; 400:687-698. [DOI: 10.1515/hsz-2018-0328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/11/2018] [Indexed: 12/11/2022]
Abstract
Abstract
The ability of cells to sense and respond to mechanical stimuli is crucial for many developmental and homeostatic processes, while mechanical dysfunction of cells has been associated with numerous pathologies including muscular dystrophies, cardiovascular defects and epithelial disorders. Yet, how cells detect and process mechanical information is still largely unclear. In this review, we outline major mechanisms underlying cellular mechanotransduction and we summarize the current understanding of how cells integrate information from distinct mechanosensitive structures to mediate complex mechanoresponses. We also discuss the concept of mechanical memory and describe how cells store information on previous mechanical events for different periods of time.
Collapse
Affiliation(s)
- Verena Kanoldt
- Group of Molecular Mechanotransduction , Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
| | - Lisa Fischer
- Group of Molecular Mechanotransduction , Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
| | - Carsten Grashoff
- Group of Molecular Mechanotransduction , Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
- Department of Quantitative Cell Biology , Institute of Molecular Cell Biology, University of Münster , 48149 Münster , Germany
| |
Collapse
|
42
|
Gates EM, LaCroix AS, Rothenberg KE, Hoffman BD. Improving Quality, Reproducibility, and Usability of FRET-Based Tension Sensors. Cytometry A 2018; 95:201-213. [PMID: 30523675 DOI: 10.1002/cyto.a.23688] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023]
Abstract
Mechanobiology, the study of how mechanical forces affect cellular behavior, is an emerging field of study that has garnered broad and significant interest. Researchers are currently seeking to better understand how mechanical signals are transmitted, detected, and integrated at a subcellular level. One tool for addressing these questions is a Förster resonance energy transfer (FRET)-based tension sensor, which enables the measurement of molecular-scale forces across proteins based on changes in emitted light. However, the reliability and reproducibility of measurements made with these sensors has not been thoroughly examined. To address these concerns, we developed numerical methods that improve the accuracy of measurements made using sensitized emission-based imaging. To establish that FRET-based tension sensors are versatile tools that provide consistent measurements, we used these methods, and demonstrated that a vinculin tension sensor is unperturbed by cell fixation, permeabilization, and immunolabeling. This suggests FRET-based tension sensors could be coupled with a variety of immuno-fluorescent labeling techniques. Additionally, as tension sensors are frequently employed in complex biological samples where large experimental repeats may be challenging, we examined how sample size affects the uncertainty of FRET measurements. In total, this work establishes guidelines to improve FRET-based tension sensor measurements, validate novel implementations of these sensors, and ensure that results are precise and reproducible. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Evan M Gates
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| | - Andrew S LaCroix
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| | - Katheryn E Rothenberg
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| |
Collapse
|
43
|
Miller PW, Pokutta S, Mitchell JM, Chodaparambil JV, Clarke DN, Nelson WJ, Weis WI, Nichols SA. Analysis of a vinculin homolog in a sponge (phylum Porifera) reveals that vertebrate-like cell adhesions emerged early in animal evolution. J Biol Chem 2018; 293:11674-11686. [PMID: 29880641 PMCID: PMC6066325 DOI: 10.1074/jbc.ra117.001325] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/21/2018] [Indexed: 01/27/2023] Open
Abstract
The evolution of cell-adhesion mechanisms in animals facilitated the assembly of organized multicellular tissues. Studies in traditional animal models have revealed two predominant adhesion structures, the adherens junction (AJ) and focal adhesions (FAs), which are involved in the attachment of neighboring cells to each other and to the secreted extracellular matrix (ECM), respectively. The AJ (containing cadherins and catenins) and FAs (comprising integrins, talin, and paxillin) differ in protein composition, but both junctions contain the actin-binding protein vinculin. The near ubiquity of these structures in animals suggests that AJ and FAs evolved early, possibly coincident with multicellularity. However, a challenge to this perspective is that previous studies of sponges-a divergent animal lineage-indicate that their tissues are organized primarily by an alternative, sponge-specific cell-adhesion mechanism called "aggregation factor." In this study, we examined the structure, biochemical properties, and tissue localization of a vinculin ortholog in the sponge Oscarella pearsei (Op). Our results indicate that Op vinculin localizes to both cell-cell and cell-ECM contacts and has biochemical and structural properties similar to those of vertebrate vinculin. We propose that Op vinculin played a role in cell adhesion and tissue organization in the last common ancestor of sponges and other animals. These findings provide compelling evidence that sponge tissues are indeed organized like epithelia in other animals and support the notion that AJ- and FA-like structures extend to the earliest periods of animal evolution.
Collapse
Affiliation(s)
| | - Sabine Pokutta
- From the Departments of Molecular and Cellular Physiology and
- Structural Biology, School of Medicine and
| | - Jennyfer M Mitchell
- the Department of Biological Sciences, University of Denver, Denver, Colorado 80208
| | - Jayanth V Chodaparambil
- From the Departments of Molecular and Cellular Physiology and
- Structural Biology, School of Medicine and
| | - D Nathaniel Clarke
- the Department of Biology, Stanford University, Stanford, California 94305 and
| | - W James Nelson
- From the Departments of Molecular and Cellular Physiology and
- the Department of Biology, Stanford University, Stanford, California 94305 and
| | - William I Weis
- From the Departments of Molecular and Cellular Physiology and
- Structural Biology, School of Medicine and
| | - Scott A Nichols
- the Department of Biological Sciences, University of Denver, Denver, Colorado 80208
| |
Collapse
|
44
|
Nicholl ID, Matsui T, Weiss TM, Stanley CB, Heller WT, Martel A, Farago B, Callaway DJE, Bu Z. α-Catenin Structure and Nanoscale Dynamics in Solution and in Complex with F-Actin. Biophys J 2018; 115:642-654. [PMID: 30037495 DOI: 10.1016/j.bpj.2018.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/17/2018] [Accepted: 07/05/2018] [Indexed: 12/26/2022] Open
Abstract
As a core component of the adherens junction, α-catenin stabilizes the cadherin/catenin complexes to the actin cytoskeleton for the mechanical coupling of cell-cell adhesion. α-catenin also modulates actin dynamics, cell polarity, and cell-migration functions that are independent of the adherens junction. We have determined the solution structures of the α-catenin monomer and dimer using in-line size-exclusion chromatography small-angle X-ray scattering, as well as the structure of α-catenin dimer in complex to F-actin filament using selective deuteration and contrast-matching small angle neutron scattering. We further present the first observation, to our knowledge, of the nanoscale dynamics of α-catenin by neutron spin-echo spectroscopy, which explicitly reveals the mobile regions of α-catenin that are crucial for binding to F-actin. In solution, the α-catenin monomer is more expanded than either protomer shown in the crystal structure dimer, with the vinculin-binding M fragment and the actin-binding domain being able to adopt different configurations. The α-catenin dimer in solution is also significantly more expanded than the dimer crystal structure, with fewer interdomain and intersubunit contacts than the crystal structure. When in complex to F-actin, the α-catenin dimer has an even more open and extended conformation than in solution, with the actin-binding domain further separated from the main body of the dimer. The α-catenin-assembled F-actin bundle develops into an ordered filament packing arrangement at increasing α-catenin/F-actin molar ratios. Together, the structural and dynamic studies reveal that α-catenin possesses dynamic molecular conformations that prime this protein to function as a mechanosensor protein.
Collapse
Affiliation(s)
- Iain D Nicholl
- Department of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom
| | - Tsutomu Matsui
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | - Thomas M Weiss
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | | | - William T Heller
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | | | | | - David J E Callaway
- Department of Chemistry and Biochemistry, City College of New York, City University of New York, New York, New York.
| | - Zimei Bu
- Department of Chemistry and Biochemistry, City College of New York, City University of New York, New York, New York.
| |
Collapse
|
45
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Cadherins in vascular smooth muscle cell (patho)biology: Quid nos scimus? Cell Signal 2018; 45:23-42. [DOI: 10.1016/j.cellsig.2018.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 12/16/2022]
|
46
|
Spiess M, Hernandez-Varas P, Oddone A, Olofsson H, Blom H, Waithe D, Lock JG, Lakadamyali M, Strömblad S. Active and inactive β1 integrins segregate into distinct nanoclusters in focal adhesions. J Cell Biol 2018; 217:1929-1940. [PMID: 29632027 PMCID: PMC5987715 DOI: 10.1083/jcb.201707075] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 02/16/2018] [Accepted: 03/26/2018] [Indexed: 01/24/2023] Open
Abstract
Through two superresolution microscopy techniques, STED and STORM, Spiess et al. visualize the organization of integrins in focal adhesions and show that active and inactive β1 integrins assemble into distinct nanoclusters within adhesions, suggesting the existence of a novel mechanism that locally coordinates integrin activity. Integrins are the core constituents of cell–matrix adhesion complexes such as focal adhesions (FAs) and play key roles in physiology and disease. Integrins fluctuate between active and inactive conformations, yet whether the activity state influences the spatial organization of integrins within FAs has remained unclear. In this study, we address this question and also ask whether integrin activity may be regulated either independently for each integrin molecule or through locally coordinated mechanisms. We used two distinct superresolution microscopy techniques, stochastic optical reconstruction microscopy (STORM) and stimulated emission depletion microscopy (STED), to visualize active versus inactive β1 integrins. We first reveal a spatial hierarchy of integrin organization with integrin molecules arranged in nanoclusters, which align to form linear substructures that in turn build FAs. Remarkably, within FAs, active and inactive β1 integrins segregate into distinct nanoclusters, with active integrin nanoclusters being more organized. This unexpected segregation indicates synchronization of integrin activities within nanoclusters, implying the existence of a coordinate mechanism of integrin activity regulation.
Collapse
Affiliation(s)
- Matthias Spiess
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Pablo Hernandez-Varas
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Wolfson Imaging Centre, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - Anna Oddone
- Institut de Ciències Fotòniques, Barcelona, Spain
| | - Helene Olofsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Hans Blom
- Science for Life Laboratory, Royal Institute of Technology, Solna, Sweden
| | - Dominic Waithe
- Wolfson Imaging Centre, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - John G Lock
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | - Staffan Strömblad
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
47
|
Coelho NM, McCulloch CA. Mechanical signaling through the discoidin domain receptor 1 plays a central role in tissue fibrosis. Cell Adh Migr 2018; 12:348-362. [PMID: 29513135 PMCID: PMC6363045 DOI: 10.1080/19336918.2018.1448353] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/20/2018] [Accepted: 02/26/2018] [Indexed: 02/08/2023] Open
Abstract
The preservation of tissue and organ architecture and function depends on tightly regulated interactions of cells with the extracellular matrix (ECM). These interactions are maintained in a dynamic equilibrium that balances intracellular, myosin-generated tension with extracellular resistance conferred by the mechanical properties of the extracellular matrix. Disturbances of this equilibrium can lead to the development of fibrotic lesions that are associated with a wide repertoire of high prevalence diseases including obstructive cardiovascular diseases, muscular dystrophy and cancer. Mechanotransduction is the process by which mechanical cues are converted into biochemical signals. At the core of mechanotransduction are sensory systems, which are frequently located at sites of cell-ECM and cell-cell contacts. As integrins (cell-ECM junctions) and cadherins (cell-cell contacts) have been extensively studied, we focus here on the properties of the discoidin domain receptor 1 (DDR1), a tyrosine kinase that mediates cell adhesion to collagen. DDR1 expression is positively associated with fibrotic lesions of heart, kidney, liver, lung and perivascular tissues. As the most common end-point of all fibrotic disorders is dysregulated collagen remodeling, we consider here the mechanical signaling functions of DDR1 in processing of fibrillar collagen that lead to tissue fibrosis.
Collapse
Affiliation(s)
- Nuno M. Coelho
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
48
|
Farley RD. Book lung development in juveniles and adults of the cobweb spider, Parasteatoda tepidariorum C. L. Koch, 1841 (Araneomorphae, Theridiidae). ARTHROPOD STRUCTURE & DEVELOPMENT 2018; 47:180-198. [PMID: 29341927 DOI: 10.1016/j.asd.2018.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/02/2018] [Accepted: 01/02/2018] [Indexed: 06/07/2023]
Abstract
Light and transmission electron microscopy were used to study the development of new book lung lamellae in juvenile and adult spiders (Parasteatoda tepidariorum). As hypothesized earlier in a study of embryos, mesenchyme cells dispersed throughout the opisthosoma (EMT) are a likely source of precursor epithelial cells (MET) for the new lamellae. The precursor cells in juveniles and adults continue many of the complex activities observed in embryos, e.g., migration, alignment, lumen formation, thinning, elongation, and secretion of the cuticle of air channel walls and trabeculae. The apicobasal polarity of precursor cells for new channels is apparently induced by the polarity pattern of precursor cells of channels produced earlier. Thus, new air and hemolymph channels extend and continue the alternating pattern of older channels. At sites more distant from the spiracle and atrium, new channels are usually produced by the mode II process (intracellular alignment and merging of vesicles). These air channels have bridging trabeculae and are quite stable in size throughout their length. At sites closer to the spiracle and atrium, new channels may be produced by mode I (coalescence of merocrine vesicle secretion). This raises the hypothesis that structural and functional differences in mode I and II channels and differing oxygen and fluid conditions with distance from the spiracle and atrium determine the mode of formation of new channels. Observations herein support an earlier hypothesis that there is some intercellular apical/apical and basal/basal affinity among the opposed surfaces of aligned precursor cells. This results in the alternating pattern of air channels at the apical and hemolymph channels at the basal cell surfaces.
Collapse
Affiliation(s)
- Roger D Farley
- Department of Biology, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
49
|
Epithelial Monolayers Coalesce on a Viscoelastic Substrate through Redistribution of Vinculin. Biophys J 2017; 113:1585-1598. [PMID: 28844472 PMCID: PMC5627150 DOI: 10.1016/j.bpj.2017.07.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/14/2017] [Accepted: 07/26/2017] [Indexed: 01/05/2023] Open
Abstract
The mechanical properties of the microenvironment play a large role in influencing cellular behavior. In particular, the tradeoff between substrate viscosity and elasticity on collective cell migration by adherent cells is highly physiologically relevant, but remains poorly understood. To investigate the specific effects of viscous substrates, we plated epithelial monolayers onto polydimethylsiloxane substrata with a range of viscosities and elasticities. We found that on viscoelastic substrates the monolayers underwent rapid and coordinated movement to generate cell-free areas. To understand the molecular mechanism of this coordinated movement, we imaged various structural and signaling proteins at cell-cell and cell-matrix junctions. Through quantitative image analysis of monolayer disruption and subcellular protein redistribution, we show that the mechanosensor protein, vinculin, is necessary and sufficient for this viscous response, during which it is lost from focal adhesions and recruited by the cadherin complex to intercellular junctions. In addition, the viscous response is dependent upon and enhanced by actomyosin contractility. Our results implicate vinculin translocation in a molecular switching mechanism that senses substrate viscoelasticity and associates with actomyosin contractility.
Collapse
|
50
|
Han MKL, van der Krogt GNM, de Rooij J. Zygotic vinculin is not essential for embryonic development in zebrafish. PLoS One 2017; 12:e0182278. [PMID: 28767718 PMCID: PMC5540497 DOI: 10.1371/journal.pone.0182278] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
The formation of multicellular tissues during development is governed by mechanical forces that drive cell shape and tissue architecture. Protein complexes at sites of adhesion to the extracellular matrix (ECM) and cell neighbors, not only transmit these mechanical forces, but also allow cells to respond to changes in force by inducing biochemical feedback pathways. Such force-induced signaling processes are termed mechanotransduction. Vinculin is a central protein in mechanotransduction that in both integrin-mediated cell-ECM and cadherin-mediated cell-cell adhesions mediates force-induced cytoskeletal remodeling and adhesion strengthening. Vinculin was found to be important for the integrity and remodeling of epithelial tissues in cell culture models and could therefore be expected to be of broad importance in epithelial morphogenesis in vivo. Besides a function in mouse heart development, however, the importance of vinculin in morphogenesis of other vertebrate tissues has remained unclear. To investigate this further, we knocked out vinculin functioning in zebrafish, which contain two fully functional isoforms designated as vinculin A and vinculin B that both show high sequence conservation with higher vertebrates. Using TALEN and CRISPR-Cas gene editing technology we generated vinculin-deficient zebrafish. While single vinculin A mutants are viable and able to reproduce, additional loss of zygotic vinculin B was lethal after embryonic stages. Remarkably, vinculin-deficient embryos do not show major developmental defects, apart from mild pericardial edemas. These results lead to the conclusion that vinculin is not of broad importance for the development and morphogenesis of zebrafish tissues.
Collapse
Affiliation(s)
- Mitchell K. L. Han
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard N. M. van der Krogt
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|