1
|
Feng X, Zhang D, Wang G, Lu L, Feng F, Wang X, Yu C, Chai Y, Zhang J, Li W, Liu J, Sun H, Yao L. Mechanisms and Therapeutic Strategies for Minority Cell-Induced Paclitaxel Resistance and Tumor Progression Mediated by Mechanical Forces. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417805. [PMID: 40270447 DOI: 10.1002/advs.202417805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Chemotherapy remains a prevalent strategy in cancer therapy; however, the emergence of drug resistance poses a considerable challenge to its efficacy. Most drug resistance arises from the accumulation of genetic mutations in a minority of resistant cells. The mechanisms underlying the emergence and progression of cancer resistance from these minority-resistant cells (MRCs) remain poorly understood. This study employs force-induced remnant magnetization spectroscopy (FIRMS) alongside various biological investigations to reveal the mechanical pathways for MRCs fostering drug resistance and tumor progression. The findings show that minority Paclitaxel-resistant cancer cells have enhanced mechanical properties. These cells can transmit high-intensity forces to surrounding sensitive cells (SCs) through the force transducer, Merlin. This force transmission facilitates the assimilation of surrounding SCs, subsequently strengthening the contraction and adhesion of tumor cells. This process is termed "mechano-assimilation," which accelerates the development of drug resistance and tumor progression. Interestingly, disturbances and reductions of mechano-assimilation within tumors can restore sensitivity to Paclitaxel both in vitro and in vivo. This study provides preliminary evidence highlighting the contribution of MRCs to the development of drug resistance and malignancy, mediated through mechanical interactions. It also establishes a foundation for future research focused on integrating mechanical factors into innovative cancer therapies.
Collapse
Affiliation(s)
- Xueyan Feng
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Di Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Guoxun Wang
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Liwei Lu
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
| | - Feng Feng
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiuyu Wang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
| | - Chanchan Yu
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yahong Chai
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jin Zhang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, 100029, P. R. China
| | - Wenchao Li
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100007, P. R. China
| | - Jing Liu
- Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Hongxia Sun
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Li Yao
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Chinese Academy of Science, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
2
|
Peña R, Baulida J. Snail1 as a key prognostic biomarker of cancer-associated fibroblasts in breast tumors. Biochim Biophys Acta Rev Cancer 2025; 1880:189316. [PMID: 40222423 DOI: 10.1016/j.bbcan.2025.189316] [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: 11/12/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
Accurate cancer diagnosis is crucial for selecting optimal treatments, yet current classification systems often include non-responders who receive ineffective therapies. Cancer-associated fibroblasts (CAFs) play a central role in tumor progression, and CAF biomarkers are increasingly recognized for their prognostic value. Recent studies have revealed significant heterogeneity within CAF populations, with distinct subtypes linked to different tumors and stages of disease. In this review, we summarize recent findings from patient samples and mouse models of breast cancer, focusing on gene signatures identified by single-cell RNA sequencing that define CAF subtypes and predict cancer prognosis. Additionally, we explore the genes and pathways regulated by Snail1, a transcription factor whose expression in breast and colon CAFs is associated with malignancy. Altogether these data emphasize the fibrotic and immunosuppressive roles of Snail1-expressing fibroblasts and unveil an undescribed streamlined Snail1-related gene signature in CAFs with prognostic potential in breast cancer and other solid tumors.
Collapse
Affiliation(s)
- Raúl Peña
- Cancer Research Program, associated unit IIBB-CSIC, Hospital del Mar Research Institute, Barcelona, Spain
| | - Josep Baulida
- Cancer Research Program, associated unit IIBB-CSIC, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
3
|
Keijzer KAE, Tsingos E, Merks RMH. How cells align to structured collagen fibrils: a hybrid cellular Potts and molecular dynamics model with dynamic mechanosensitive focal adhesions. Front Cell Dev Biol 2025; 12:1462277. [PMID: 39834385 PMCID: PMC11743931 DOI: 10.3389/fcell.2024.1462277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Many mammalian cells, including endothelial cells and fibroblasts, align and elongate along the orientation of extracellular matrix (ECM) fibers in a gel when cultured in vitro. During cell elongation, clusters of focal adhesions (FAs) form near the poles of the elongating cells. FAs are mechanosensitive clusters of adhesions that grow under mechanical tension exerted by the cells' pulling on the ECM and shrink when the tension is released. In this study, we use mathematical modeling to study the hypothesis that mechanical reciprocity between cells and the ECM is sufficient for directing cell shape changes and orientation. We show that FAs are preferentially stabilized along the orientation of ECM fibers, where cells can generate higher tension than in directions perpendicular to the ECM fibers. We present a hybrid computational model coupling three mathematical approaches: first, the cellular Potts model (CPM) describes an individual contractile cell; second, molecular dynamics (MD) represent the ECM as a network of cross-linked, deformable fibers; third, a set of ordinary differential equations (ODEs) describes the dynamics of the cell's FAs, in terms of a balance between assembly and a mechanoresponsive disassembly. The resulting computational model shows that mechanical reciprocity suffices for stiffness-dependent cell spreading, local ECM remodeling, and ECM-alignment-dependent cell elongation. These combined effects are sufficient to explain how cell morphology is influenced by the local ECM structure and mechanics.
Collapse
Affiliation(s)
- Koen A. E. Keijzer
- Mathematical Institute, Faculty of Science, Leiden University, Leiden, Netherlands
| | - Erika Tsingos
- Mathematical Institute, Faculty of Science, Leiden University, Leiden, Netherlands
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, Netherlands
| | - Roeland M. H. Merks
- Mathematical Institute, Faculty of Science, Leiden University, Leiden, Netherlands
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, Netherlands
| |
Collapse
|
4
|
Gasser E, Su E, Vaidžiulytė K, Abbade N, Cognart H, Manneville JB, Viovy JL, Piel M, Pierga JY, Terao K, Villard C. Deformation under flow and morphological recovery of cancer cells. LAB ON A CHIP 2024; 24:3930-3944. [PMID: 38993177 PMCID: PMC11302772 DOI: 10.1039/d4lc00246f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Abstract
The metastatic cascade includes a blood circulation step for cells detached from the primary tumor. This stage involves significant shear stress as well as large and fast deformation as the cells circulate through the microvasculature. These mechanical stimuli are well reproduced in microfluidic devices. However, the recovery dynamics after deformation is also pivotal to understand how a cell can pass through the multiple capillary constrictions encountered during a single hemodynamic cycle. The microfluidic system developed in this work allows single cell recovery to be studied under flow-free conditions following pressure-actuated cell deformation inside constricted microchannels. We used three breast cancer cell lines - namely MCF-7, SK-BR3 and MDA-MB231 - as cellular models representative of different cancer phenotypes. Changing the size of the constriction allows exploration of moderate to strong deformation regimes, the latter being associated with the formation of plasma membrane blebs. In the regime of moderate deformation, all cell types display a fast elastic recovery behavior followed by a slower viscoelastic regime, well described by a double exponential decay. Among the three cell types, cells of the mesenchymal phenotype, i.e. the MDA-MB231 cells, are softer and the most fluid-like, in agreement with previous studies. Our main finding here is that the fast elastic recovery regime revealed by our novel microfluidic system is under the control of cell contractility ensured by the integrity of the cell cortex. Our results suggest that the cell cortex plays a major role in the transit of circulating tumor cells by allowing their fast morphological recovery after deformation in blood capillaries.
Collapse
Affiliation(s)
- Emile Gasser
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
| | - Emilie Su
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
- Laboratoire Matière et Systèmes Complexes (MSC), CNRS UMR 7057, Université Paris Cité, 10 Rue Alice Domon et Léonie Duquet, F-75013 Paris, France
| | - Kotryna Vaidžiulytė
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Nassiba Abbade
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Hamizah Cognart
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
| | - Jean-Baptiste Manneville
- Laboratoire Matière et Systèmes Complexes (MSC), CNRS UMR 7057, Université Paris Cité, 10 Rue Alice Domon et Léonie Duquet, F-75013 Paris, France
| | - Jean-Louis Viovy
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Jean-Yves Pierga
- Département d'Oncologie Médicale de l'Institut Curie et Université Paris Cité, France
| | - Kyohei Terao
- Nano-Micro Structure Device Integrated Research Center, Kagawa University, 2217-20 Hayashi-cho, Takamatsu 761-0396, Japan.
| | - Catherine Villard
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
| |
Collapse
|
5
|
Tsingos E, Bakker BH, Keijzer KAE, Hupkes HJ, Merks RMH. Hybrid cellular Potts and bead-spring modeling of cells in fibrous extracellular matrix. Biophys J 2023; 122:2609-2622. [PMID: 37183398 PMCID: PMC10397577 DOI: 10.1016/j.bpj.2023.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023] Open
Abstract
The mechanical interaction between cells and the extracellular matrix (ECM) is fundamental to coordinate collective cell behavior in tissues. Relating individual cell-level mechanics to tissue-scale collective behavior is a challenge that cell-based models such as the cellular Potts model (CPM) are well-positioned to address. These models generally represent the ECM with mean-field approaches, which assume substrate homogeneity. This assumption breaks down with fibrous ECM, which has nontrivial structure and mechanics. Here, we extend the CPM with a bead-spring model of ECM fiber networks modeled using molecular dynamics. We model a contractile cell pulling with discrete focal adhesion-like sites on the fiber network and demonstrate agreement with experimental spatiotemporal fiber densification and displacement. We show that at high network cross-linking, contractile cell forces propagate over at least eight cell diameters, decaying with distance with power law exponent n= 0.35 - 0.65 typical of viscoelastic ECMs. Further, we use in silico atomic force microscopy to measure local cell-induced network stiffening consistent with experiments. Our model lays the foundation for investigating how local and long-ranged cell-ECM mechanobiology contributes to multicellular morphogenesis.
Collapse
Affiliation(s)
- Erika Tsingos
- Mathematical Institute, Leiden University, Leiden, the Netherlands.
| | | | - Koen A E Keijzer
- Mathematical Institute, Leiden University, Leiden, the Netherlands
| | | | - Roeland M H Merks
- Mathematical Institute, Leiden University, Leiden, the Netherlands; Institute for Biology Leiden, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
6
|
Smith SJ, Guillon E, Holley SA. The roles of inter-tissue adhesion in development and morphological evolution. J Cell Sci 2022; 135:275268. [PMID: 35522159 PMCID: PMC9264361 DOI: 10.1242/jcs.259579] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The study of how neighboring tissues physically interact with each other, inter-tissue adhesion, is an emerging field at the interface of cell biology, biophysics and developmental biology. Inter-tissue adhesion can be mediated by either cell-extracellular matrix adhesion or cell-cell adhesion, and both the mechanisms and consequences of inter-tissue adhesion have been studied in vivo in numerous vertebrate and invertebrate species. In this Review, we discuss recent progress in understanding the many functions of inter-tissue adhesion in development and evolution. Inter-tissue adhesion can couple the motion of adjacent tissues, be the source of mechanical resistance that constrains morphogenesis, and transmit tension required for normal development. Tissue-tissue adhesion can also create mechanical instability that leads to tissue folding or looping. Transient inter-tissue adhesion can facilitate tissue invasion, and weak tissue adhesion can generate friction that shapes and positions tissues within the embryo. Lastly, we review studies that reveal how inter-tissue adhesion contributes to the diversification of animal morphologies.
Collapse
Affiliation(s)
- Sarah Jacquelyn Smith
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Emilie Guillon
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
7
|
Cell Chirality Regulates Coherent Angular Motion on Small Circular Substrates. Biophys J 2022; 121:1931-1939. [DOI: 10.1016/j.bpj.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/01/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
|
8
|
Transcriptomic Analysis of Fish Hosts Responses to Nervous Necrosis Virus. Pathogens 2022; 11:pathogens11020201. [PMID: 35215144 PMCID: PMC8875540 DOI: 10.3390/pathogens11020201] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 11/24/2022] Open
Abstract
Nervous necrosis virus (NNV) has been responsible for mass mortalities in the aquaculture industry worldwide, with great economic and environmental impact. The present review aims to summarize the current knowledge of gene expression responses to nervous necrosis virus infection in different fish species based on transcriptomic analysis data. Four electronic databases, including PubMed, Web of Science, and SCOPUS were searched, and more than 500 publications on the subject were identified. Following the application of the appropriate testing, a total of 24 articles proved eligible for this review. NNV infection of different host species, in different developmental stages and tissues, presented in the eligible publications, are described in detail, revealing and highlighting genes and pathways that are most affected by the viral infection. Those transcriptome studies of NNV infected fish are oriented in elucidating the roles of genes/biomarkers for functions of special interest, depending on each study’s specific emphasis. This review presents a first attempt to provide an overview of universal host reaction mechanisms to viral infections, which will provide us with new perspectives to overcome NNV infection to build healthier and sustainable aquaculture systems.
Collapse
|
9
|
Fang C, Yao J, Zhang Y, Lin Y. Active chemo-mechanical feedbacks dictate the collective migration of cells on patterned surfaces. Biophys J 2022; 121:1266-1275. [PMID: 35183521 PMCID: PMC9034249 DOI: 10.1016/j.bpj.2022.02.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/13/2021] [Accepted: 02/15/2022] [Indexed: 11/02/2022] Open
Abstract
Recent evidence has demonstrated that, when cultured on micro-patterned surfaces, living cells can move in a coordinated manner and form distinct migration patterns, including flowing chain, suspended propagating bridge, rotating vortex, etc. However, the fundamental question of exactly how and why cells migrate in these fashions remains elusive. Here, we present a theoretical investigation to show that the tight interplay between internal cellular activities, such as chemo-mechanical feedbacks and polarization, and external geometrical constraints are behind these intriguing experimental observations. In particular, on narrow strip patterns, strongly force-dependent cellular contractility and intercellular adhesion were found to be critical for reinforcing the leading edge of the migrating cell monolayer and eventually result in the formation of suspended cell bridges flying over nonadhesive regions. On the other hand, a weak force-contractility feedback led to the movement of cells like a flowing chain along the adhesive strip. Finally, we also showed that the random polarity forces generated in migrating cells are responsible for driving them into rotating vortices on strips with width above a threshold value (~10 times the size of the cell).
Collapse
|
10
|
Engel-Pizcueta C, Pujades C. Interplay Between Notch and YAP/TAZ Pathways in the Regulation of Cell Fate During Embryo Development. Front Cell Dev Biol 2021; 9:711531. [PMID: 34490262 PMCID: PMC8417249 DOI: 10.3389/fcell.2021.711531] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/02/2021] [Indexed: 12/23/2022] Open
Abstract
Cells in growing tissues receive both biochemical and physical cues from their microenvironment. Growing evidence has shown that mechanical signals are fundamental regulators of cell behavior. However, how physical properties of the microenvironment are transduced into critical cell behaviors, such as proliferation, progenitor maintenance, or differentiation during development, is still poorly understood. The transcriptional co-activators YAP/TAZ shuttle between the cytoplasm and the nucleus in response to multiple inputs and have emerged as important regulators of tissue growth and regeneration. YAP/TAZ sense and transduce physical cues, such as those from the extracellular matrix or the actomyosin cytoskeleton, to regulate gene expression, thus allowing them to function as gatekeepers of progenitor behavior in several developmental contexts. The Notch pathway is a key signaling pathway that controls binary cell fate decisions through cell-cell communication in a context-dependent manner. Recent reports now suggest that the crosstalk between these two pathways is critical for maintaining the balance between progenitor maintenance and cell differentiation in different tissues. How this crosstalk integrates with morphogenesis and changes in tissue architecture during development is still an open question. Here, we discuss how progenitor cell proliferation, specification, and differentiation are coordinated with morphogenesis to construct a functional organ. We will pay special attention to the interplay between YAP/TAZ and Notch signaling pathways in determining cell fate decisions and discuss whether this represents a general mechanism of regulating cell fate during development. We will focus on research carried out in vertebrate embryos that demonstrate the important roles of mechanical cues in stem cell biology and discuss future challenges.
Collapse
Affiliation(s)
- Carolyn Engel-Pizcueta
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
11
|
Blackley DG, Cooper JH, Pokorska P, Ratheesh A. Mechanics of developmental migration. Semin Cell Dev Biol 2021; 120:66-74. [PMID: 34275746 DOI: 10.1016/j.semcdb.2021.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 02/01/2023]
Abstract
The ability to migrate is a fundamental property of animal cells which is essential for development, homeostasis and disease progression. Migrating cells sense and respond to biochemical and mechanical cues by rapidly modifying their intrinsic repertoire of signalling molecules and by altering their force generating and transducing machinery. We have a wealth of information about the chemical cues and signalling responses that cells use during migration. Our understanding of the role of forces in cell migration is rapidly evolving but is still best understood in the context of cells migrating in 2D and 3D environments in vitro. Advances in live imaging of developing embryos combined with the use of experimental and theoretical tools to quantify and analyse forces in vivo, has begun to shed light on the role of mechanics in driving embryonic cell migration. In this review, we focus on the recent studies uncovering the physical basis of embryonic cell migration in vivo. We look at the physical basis of the classical steps of cell migration such as protrusion formation and cell body translocation and review the recent research on how these processes work in the complex 3D microenvironment of a developing organism.
Collapse
Affiliation(s)
- Deannah G Blackley
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Jack H Cooper
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Paulina Pokorska
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Aparna Ratheesh
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
12
|
Inman A, Smutny M. Feeling the force: Multiscale force sensing and transduction at the cell-cell interface. Semin Cell Dev Biol 2021; 120:53-65. [PMID: 34238674 DOI: 10.1016/j.semcdb.2021.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022]
Abstract
A universal principle of all living cells is the ability to sense and respond to mechanical stimuli which is essential for many biological processes. Recent efforts have identified critical mechanosensitive molecules and response pathways involved in mechanotransduction during development and tissue homeostasis. Tissue-wide force transmission and local force sensing need to be spatiotemporally coordinated to precisely regulate essential processes during development such as tissue morphogenesis, patterning, cell migration and organogenesis. Understanding how cells identify and interpret extrinsic forces and integrate a specific response on cell and tissue level remains a major challenge. In this review we consider important cellular and physical factors in control of cell-cell mechanotransduction and discuss their significance for cell and developmental processes. We further highlight mechanosensitive macromolecules that are known to respond to external forces and present examples of how force responses can be integrated into cell and developmental programs.
Collapse
Affiliation(s)
- Angus Inman
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK
| | - Michael Smutny
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK.
| |
Collapse
|
13
|
Abdollahiyan P, Oroojalian F, Baradaran B, de la Guardia M, Mokhtarzadeh A. Advanced mechanotherapy: Biotensegrity for governing metastatic tumor cell fate via modulating the extracellular matrix. J Control Release 2021; 335:596-618. [PMID: 34097925 DOI: 10.1016/j.jconrel.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Mechano-transduction is the procedure of mechanical stimulus translation via cells, among substrate shear flow, topography, and stiffness into a biochemical answer. TAZ and YAP are transcriptional coactivators which are recognized as relay proteins that promote mechano-transduction within the Hippo pathway. With regard to healthy cells in homeostasis, mechano-transduction regularly restricts proliferation, and TAZ and YAP are totally inactive. During cancer development a YAP/TAZ - stimulating positive response loop is formed between the growing tumor and the stiffening ECM. As tumor developments, local stromal and cancerous cells take advantage of mechanotransduction to enhance proliferation, induce their migratory into remote tissues, and promote chemotherapeutic resistance. As a newly progresses paradigm, nanoparticle-conjunctions (such as magnetic nanoparticles, and graphene derivatives nanoparticles) hold significant promises for remote regulation of cells and their relevant events at molecular scale. Despite outstanding developments in employing nanoparticles for drug targeting studies, the role of nanoparticles on cellular behaviors (proliferation, migration, and differentiation) has still required more evaluations in the field of mechanotherapy. In this paper, the in-depth contribution of mechano-transduction is discussed during tumor progression, and how these consequences can be evaluated in vitro.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Valencia, Spain
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
14
|
Aoun L, Nègre P, Gonsales C, Seveau de Noray V, Brustlein S, Biarnes-Pelicot M, Valignat MP, Theodoly O. Leukocyte transmigration and longitudinal forward-thrusting force in a microfluidic Transwell device. Biophys J 2021; 120:2205-2221. [PMID: 33838136 DOI: 10.1016/j.bpj.2021.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 01/21/2023] Open
Abstract
Transmigration of leukocytes across blood vessels walls is a critical step of the immune response. Transwell assays examine transmigration properties in vitro by counting cells passages through a membrane; however, the difficulty of in situ imaging hampers a clear disentanglement of the roles of adhesion, chemokinesis, and chemotaxis. We used here microfluidic Transwells to image the cells' transition from 2D migration on a surface to 3D migration in a confining microchannel and measure cells longitudinal forward-thrusting force in microchannels. Primary human effector T lymphocytes adhering with integrins LFA-1 (αLβ2) had a marked propensity to transmigrate in Transwells without chemotactic cue. Both adhesion and contractility were important to overcome the critical step of nucleus penetration but were remarkably dispensable for 3D migration in smooth microchannels deprived of topographic features. Transmigration in smooth channels was qualitatively consistent with a propulsion by treadmilling of cell envelope and squeezing of cell trailing edge. Stalling conditions of 3D migration were then assessed by imposing pressure drops across microchannels. Without specific adhesion, the cells slid backward with subnanonewton forces, showing that 3D migration under stress is strongly limited by a lack of adhesion and friction with channels. With specific LFA-1 mediated adhesion, stalling occurred at around 3 and 6 nN in 2 × 4 and 4 × 4 μm2 channels, respectively, supporting that stalling of adherent cells was under pressure control rather than force control. The stall pressure of 4 mbar is consistent with the pressure of actin filament polymerization that mediates lamellipod growth. The arrest of adherent cells under stress therefore seems controlled by the compression of the cell leading edge, which perturbs cells front-rear polarization and triggers adhesion failure or polarization reversal. Although stalling assays in microfluidic Transwells do not mimic in vivo transmigration, they provide a powerful tool to scrutinize 2D and 3D migration, barotaxis, and chemotaxis.
Collapse
Affiliation(s)
- Laurene Aoun
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Paulin Nègre
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Cristina Gonsales
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Sophie Brustlein
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Marie-Pierre Valignat
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Olivier Theodoly
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
15
|
Chen X, Li Y, Yao T, Jia R. Benefits of Zebrafish Xenograft Models in Cancer Research. Front Cell Dev Biol 2021; 9:616551. [PMID: 33644052 PMCID: PMC7905065 DOI: 10.3389/fcell.2021.616551] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
As a promising in vivo tool for cancer research, zebrafish have been widely applied in various tumor studies. The zebrafish xenograft model is a low-cost, high-throughput tool for cancer research that can be established quickly and requires only a small sample size, which makes it favorite among researchers. Zebrafish patient-derived xenograft (zPDX) models provide promising evidence for short-term clinical treatment. In this review, we discuss the characteristics and advantages of zebrafish, such as their transparent and translucent features, the use of vascular fluorescence imaging, the establishment of metastatic and intracranial orthotopic models, individual pharmacokinetics measurements, and tumor microenvironment. Furthermore, we introduce how these characteristics and advantages are applied other in tumor studies. Finally, we discuss the future direction of the use of zebrafish in tumor studies and provide new ideas for the application of it.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tengteng Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
16
|
Reyes Lua AM, Tonnicchia S, Giampietro C, Mazza E, Ferrari A. Evaluation of Chemo- and Photo-toxicity of a Live Fluorescent Dye for Cell Analysis. Photochem Photobiol 2020; 97:448-452. [PMID: 33277719 DOI: 10.1111/php.13362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 11/30/2020] [Indexed: 12/01/2022]
Abstract
Live cell imaging is used to track the dynamic adaptation of cell size and motility to various external factors. Bright-field configuration can be used for these experiments; however, the analysis can be challenging and difficult to automate. In this direction, a superior alternative is represented by the use of live cell dyes, which provide intense fluorescence from subcellular structures of living cells. Yet, the potential chemo- and photo-toxicity of the fluorophores poses the necessity of an accurate protocol optimization to avoid artefacts. Toxicity studies generally focus on cell proliferation and apoptosis, neglecting the cellular activities under investigation. Here, we present the case of SYTO 13 in combination with primary endothelial cells. The optimization of the staining procedure is tested comparing cell proliferation and motility rate. In addition, the combined effect of staining and fluorescent illumination, reporting for photochemical toxicity, is evaluated. We demonstrate that while cell viability and proliferation are mainly unaffected by the staining and imagining protocols, a significant reduction of the motility rate is induced both by the chemical dye alone and in combination with fluorescent illumination. The general implications for this procedure are discussed.
Collapse
Affiliation(s)
| | - Simone Tonnicchia
- Institute for Mechanical Systems, ETH Zurich, Zürich, Switzerland.,Swiss Federal Laboratories for Material Science and Technologies, EMPA, Dübendorf, Switzerland
| | - Costanza Giampietro
- Swiss Federal Laboratories for Material Science and Technologies, EMPA, Dübendorf, Switzerland
| | - Edoardo Mazza
- Institute for Mechanical Systems, ETH Zurich, Zürich, Switzerland.,Swiss Federal Laboratories for Material Science and Technologies, EMPA, Dübendorf, Switzerland
| | - Aldo Ferrari
- Institute for Mechanical Systems, ETH Zurich, Zürich, Switzerland.,Swiss Federal Laboratories for Material Science and Technologies, EMPA, Dübendorf, Switzerland
| |
Collapse
|
17
|
Strickfaden H. Reflections on the organization and the physical state of chromatin in eukaryotic cells. Genome 2020; 64:311-325. [PMID: 33306433 DOI: 10.1139/gen-2020-0132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In recent years, our perception of chromatin structure and organization in the cell nucleus has changed in fundamental ways. The 30 nm chromatin fiber has lost its status as an essential in vivo structure. Hi-C and related biochemical methods, advanced electron and super-resolved fluorescence microscopy, together with concepts from soft matter physics, have revolutionized the field. A comprehensive understanding of the structural and functional interactions that regulate cell cycle and cell type specific nuclear functions appears within reach, but it requires the integration of top-down and bottom-up approachs. In this review, I present an update on nuclear architecture studies with an emphasis on organization and the controversy regarding the physical state of chromatin in cells.
Collapse
Affiliation(s)
- Hilmar Strickfaden
- Departments of Cell Biology and Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Departments of Cell Biology and Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
Dynamic optima in cell sizes during early development enable normal gastrulation in zebrafish embryos. Dev Biol 2020; 468:26-40. [DOI: 10.1016/j.ydbio.2020.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/21/2022]
|
19
|
Sumbal J, Belisova D, Koledova Z. Fibroblasts: The grey eminence of mammary gland development. Semin Cell Dev Biol 2020; 114:134-142. [PMID: 33158729 DOI: 10.1016/j.semcdb.2020.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/21/2020] [Accepted: 10/25/2020] [Indexed: 02/03/2023]
Abstract
The essential role of mammary gland stroma in the regulation of mammary epithelial development, function, and cancer has long been recognized. Only recently, though, the functions of individual stromal cell populations have begun to become more clarified. Mammary fibroblasts have emerged as master regulators and modulators of epithelial cell behavior through paracrine signaling, extracellular matrix production and remodeling, and through regulation of other stromal cell types. In this review article, we summarize the crucial studies that helped to untangle the roles of fibroblasts in mammary gland development. Furthermore, we discuss the origin, heterogeneity, and plasticity of mammary fibroblasts during mammary development and cancer progression.
Collapse
Affiliation(s)
- Jakub Sumbal
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Denisa Belisova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
20
|
Korla PK, Chen CC, Gracilla DE, Lai MT, Chen CM, Chen HY, Hwang T, Chen SY, Sheu JJC. Somatic mutational landscapes of adherens junctions and their functional consequences in cutaneous melanoma development. Theranostics 2020; 10:12026-12043. [PMID: 33204327 PMCID: PMC7667680 DOI: 10.7150/thno.46705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023] Open
Abstract
Cell-cell interaction in skin homeostasis is tightly controlled by adherens junctions (AJs). Alterations in such regulation lead to melanoma development. However, mutations in AJs and their functional consequences are still largely unknown. Methods: Cadherin mutations in skin cutaneous melanoma were identified using sequencing data from TCGA dataset, followed by cross-validation with data from non-TCGA cohorts. Mutations with significant occurrence were subjected to structural prediction using MODELLER and functional protein simulation using GROMACS software. Neo-antigen prediction was carried out using NetMHCpan tool. Cell-based fluorescence reporter assay was used to validate β-catenin activity in the presence of cadherin mutations. Clinical significance was analyzed using datasets from TCGA and other non-TCGA cohorts. Targeted gene exon sequencing and immunofluorescence staining on melanoma tissues were performed to confirm the in silico findings. Results: Highly frequent mutations in type-II classical cadherins were found in melanoma with one unique recurrent mutation (S524L) in the fifth domain of CDH6, which potentially destabilizes Ca2+-binding and cell-cell contacts. Mutational co-occurrence and physical dynamics analyses placed CDH6 at the center of the top-four mutated cadherins (core CDHs; all type-II), suggesting altered heterophilic interactions in melanoma development. Mutations in the intracellular domains significantly disturbed CDH6/β-catenin complex formation, resulting in β-catenin translocation into cytosol or nucleus and dysregulation of canonical Wnt/β-catenin signaling. Although mutations in core CDH genes correlated with advanced cancer stages and lymph node invasion, the overall and disease-free survival times in those patients were longer in patients with wild-type. Peptide/MHC-I binding affinity predictions confirmed overall increased neo-antigen potentials of mutated cadherins, which associated with T-lymphocyte infiltration and better clinical outcomes after immunotherapy. Conclusion: Changes in cell-cell communications by somatic mutations in AJ cadherins function as one of mechanisms to trigger melanoma development. Certain mutations in AJs may serve as potential neo-antigens which conversely benefit patients for longer survival times.
Collapse
|
21
|
Sun J, Macabenta F, Akos Z, Stathopoulos A. Collective Migrations of Drosophila Embryonic Trunk and Caudal Mesoderm-Derived Muscle Precursor Cells. Genetics 2020; 215:297-322. [PMID: 32487692 PMCID: PMC7268997 DOI: 10.1534/genetics.120.303258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 04/17/2020] [Indexed: 01/06/2023] Open
Abstract
Mesoderm migration in the Drosophila embryo is a highly conserved, complex process that is required for the formation of specialized tissues and organs, including the somatic and visceral musculature. In this FlyBook chapter, we will compare and contrast the specification and migration of cells originating from the trunk and caudal mesoderm. Both cell types engage in collective migrations that enable cells to achieve new positions within developing embryos and form distinct tissues. To start, we will discuss specification and early morphogenetic movements of the presumptive mesoderm, then focus on the coordinate movements of the two subtypes trunk mesoderm and caudal visceral mesoderm, ending with a comparison of these processes including general insights gained through study.
Collapse
Affiliation(s)
- Jingjing Sun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Frank Macabenta
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Zsuzsa Akos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Angelike Stathopoulos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
22
|
Connolly S, Newport D, McGourty K. The mechanical responses of advecting cells in confined flow. BIOMICROFLUIDICS 2020; 14:031501. [PMID: 32454924 PMCID: PMC7200165 DOI: 10.1063/5.0005154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/21/2020] [Indexed: 05/03/2023]
Abstract
Fluid dynamics have long influenced cells in suspension. Red blood cells and white blood cells are advected through biological microchannels in both the cardiovascular and lymphatic systems and, as a result, are subject to a wide variety of complex fluidic forces as they pass through. In vivo, microfluidic forces influence different biological processes such as the spreading of infection, cancer metastasis, and cell viability, highlighting the importance of fluid dynamics in the blood and lymphatic vessels. This suggests that in vitro devices carrying cell suspensions may influence the viability and functionality of cells. Lab-on-a-chip, flow cytometry, and cell therapies involve cell suspensions flowing through microchannels of approximately 100-800 μ m. This review begins by examining the current fundamental theories and techniques behind the fluidic forces and inertial focusing acting on cells in suspension, before exploring studies that have investigated how these fluidic forces affect the reactions of suspended cells. In light of these studies' findings, both in vivo and in vitro fluidic cell microenvironments shall also be discussed before concluding with recommendations for the field.
Collapse
Affiliation(s)
- S Connolly
- School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | - D Newport
- School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | | |
Collapse
|
23
|
Ge H, Lin K, Zhou C, Lin Q, Zhang Z, Wu J, Zheng L, Yang Q, Wu S, Chen W, Wang Y. A multi-omic analysis of orange-spotted grouper larvae infected with nervous necrosis virus identifies increased adhesion molecules and collagen synthesis in the persistent state. FISH & SHELLFISH IMMUNOLOGY 2020; 98:595-604. [PMID: 32004615 DOI: 10.1016/j.fsi.2020.01.056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 06/10/2023]
Abstract
Grouper (Epinephelus coioides) is an important commercial maricultural fish, which suffers from nervous necrosis virus (NNV) infection. The molecular mechanisms underlying the pathogenesis of the viral infection are not clear. In this study, we combined deep RNA sequencing and label-free mass spectrum for the first time to analyze the transcriptomic and proteomic profiles in infected/dead, infected/survival (persistent), and infection-free (control)orange-spotted groupers in the larval stage. Further analyses showed that the transcriptome and proteome changed dramatically among the three distinct groups, especially differentially-expressed genes in the infected/dead and infected/survival larvae enriched for pathways related to immune response. Notably, the overlapped genes between transcriptomes and proteomes identified that genes related to collagen synthesis and adhesion molecules were enhanced in the persistent (infected/survival) stage, which might contribute to suppressing the acute and lethal immune responses upon NNV infection. These transcriptomic and proteomic datasets enable the investigation of molecular mechanisms underlying NNV infection, thus may help further development of molecular breeding in marine fishery.
Collapse
Affiliation(s)
- Hui Ge
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China
| | - Kebing Lin
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China
| | - Chen Zhou
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China.
| | - Qi Lin
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China
| | - Ziping Zhang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350117, China
| | - Jianshao Wu
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China
| | - Leyun Zheng
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China
| | - Qiuhua Yang
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China
| | - Shuiqing Wu
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen, 361012, China
| | - Wei Chen
- Shanghai Applied Protein Technology Co., Ltd, China
| | - Yilei Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, 361021, China.
| |
Collapse
|
24
|
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
|
25
|
Finegan TM, Bergstralh DT. Division orientation: disentangling shape and mechanical forces. Cell Cycle 2019; 18:1187-1198. [PMID: 31068057 PMCID: PMC6592245 DOI: 10.1080/15384101.2019.1617006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022] Open
Abstract
Oriented cell divisions are essential for the generation of cell diversity and for tissue shaping during morphogenesis. Cells in tissues are mechanically linked to their neighbors, upon which they impose, and from which they experience, physical force. Recent work in multiple systems has revealed that tissue-level physical forces can influence the orientation of cell division. A long-standing question is whether forces are communicated to the spindle orienting machinery via cell shape or directly via mechanosensing intracellular machinery. In this article, we review the current evidence from diverse model systems that show spindles are oriented by tissue-level physical forces and evaluate current models and molecular mechanisms proposed to explain how the spindle orientation machinery responds to extrinsic force.
Collapse
Affiliation(s)
- Tara M. Finegan
- Department of Biology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
26
|
The knocking down of the oncoprotein Golgi phosphoprotein 3 in T98G cells of glioblastoma multiforme disrupts cell migration by affecting focal adhesion dynamics in a focal adhesion kinase-dependent manner. PLoS One 2019; 14:e0212321. [PMID: 30779783 PMCID: PMC6380552 DOI: 10.1371/journal.pone.0212321] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 01/31/2019] [Indexed: 01/29/2023] Open
Abstract
Golgi phosphoprotein 3 (GOLPH3) is a conserved protein of the Golgi apparatus that in humans has been implicated in tumorigenesis. However, the precise function of GOLPH3 in malignant transformation is still unknown. Nevertheless, clinicopathological data shows that in more than a dozen kinds of cancer, including gliomas, GOLPH3 could be found overexpressed, which correlates with poor prognosis. Experimental data shows that overexpression of GOLPH3 leads to transformation of primary cells and to tumor growth enhancement. Conversely, the knocking down of GOLPH3 in GOLPH3-overexpressing tumor cells reduces tumorigenic features, such as cell proliferation and cell migration and invasion. The cumulative evidence indicate that GOLPH3 is an oncoprotein that promotes tumorigenicity by a mechanism that impact at different levels in different types of cells, including the sorting of Golgi glycosyltransferases, signaling pathways, and the actin cytoskeleton. How GOLPH3 connects mechanistically these processes has not been determined yet. Further studies are important to have a more complete understanding of the role of GOLPH3 as oncoprotein. Given the genetic diversity in cancer, a still outstanding aspect is how in this inherent heterogeneity GOLPH3 could possibly exert its oncogenic function. We have aimed to evaluate the contribution of GOLPH3 overexpression in the malignant phenotype of different types of tumor cells. Here, we analyzed the effect on cell migration that resulted from stable, RNAi-mediated knocking down of GOLPH3 in T98G cells of glioblastoma multiforme, a human glioma cell line with unique features. We found that the reduction of GOLPH3 levels produced dramatic changes in cell morphology, involving rearrangements of the actin cytoskeleton and reduction in the number and dynamics of focal adhesions. These effects correlated with decreased cell migration and invasion due to affected persistence and directionality of cell motility. Moreover, the knocking down of GOLPH3 also caused a reduction in autoactivation of focal adhesion kinase (FAK), a cytoplasmic tyrosine kinase that regulates focal adhesions. Our data support a model in which GOLPH3 in T98G cells promotes cell migration by stimulating the activity of FAK.
Collapse
|