1
|
Fajdiga L, Zemljič Š, Kokalj T, Derganc J. Shear flow deformability cytometry: A microfluidic method advancing towards clinical use - A review. Anal Chim Acta 2025; 1355:343894. [PMID: 40274322 DOI: 10.1016/j.aca.2025.343894] [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: 12/12/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Shear flow deformability cytometry is an emerging microfluidic technique that has undergone significant advances in the last few years and offers considerable potential for clinical diagnostics and disease monitoring. By simultaneously measuring mechanical and morphological parameters of single cells, it offers a comprehensive extension of traditional cell analysis, delivering unique insight into cell deformability, which is gaining recognition as a novel biomarker for health and disease. Due to its operating principle, the method is particularly suitable for the clinical analysis of blood samples. RESULTS This review focuses on the recent developments in shear flow deformability cytometry, which is a widely adopted variant of deformability cytometry. It has a strong potential for applications in clinical practice due to its robust and simple operation, demonstrated applications with whole blood samples, as well as its high throughput, which can reach approximately 1000 cells per second. We begin by discussing some basic factors that influence the mechanical properties of cells and give an overview of deformability cytometry and its operational principles for samples from blood, cultured cells and tissues. Next, we review recent clinically relevant applications in analysis of blood and cancer cells. Finally, we address key challenges to clinical adoption, such as regulatory approval, scalable manufacturing, and workflow integration, emphasizing the need for further validation studies to facilitate clinical implementation. SIGNIFICANCE This article uniquely emphasizes the clinical relevance of microfluidic shear flow deformability cytometry, by giving an overview of mechanical and morphological biomarkers studied in clinically significant samples. In addition, it addresses critical barriers to clinical translation. By identifying these obstacles, this article aims to demonstrate the potential of deformability cytometry to bridge the gap between the research and the routine medical practice.
Collapse
Affiliation(s)
- Lija Fajdiga
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Špela Zemljič
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Tadej Kokalj
- Institute of Metals and Technology, Lepi pot 11, 1000, Ljubljana, Slovenia.
| | - Jure Derganc
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Montemurro M, Monier B, Suzanne M. The mechanical state of pre-tumoral epithelia controls subsequent Drosophila tumor aggressiveness. Dev Cell 2025; 60:1036-1052.e7. [PMID: 39765232 DOI: 10.1016/j.devcel.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/13/2024] [Accepted: 12/04/2024] [Indexed: 04/10/2025]
Abstract
Tumors evolve through the acquisition of increasingly aggressive traits associated with dysplasia. This progression is accompanied by alterations in tumor mechanical properties, especially through extracellular matrix remodeling. However, the contribution of pre-tumoral tissue mechanics to tumor aggressiveness remains poorly known in vivo. Here, we show that adherens junction tension in pre-tumoral tissues dictates subsequent tumor evolution in Drosophila. Increased cell contractility, observed in aggressive tumors before any sign of tissue overgrowth, proved sufficient to trigger dysplasia in normally hyperplastic tumors. In addition, high contractility precedes any changes in cell polarity and contributes to tumor evolution through cell death induction, which favors cell-cell junction weakening. Overall, our results highlight the need to re-evaluate the roles of tumoral cell death and identify pre-tumoral cell mechanics as an unsuspected early marker and key trigger of tumor aggressiveness.
Collapse
Affiliation(s)
- Marianne Montemurro
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France
| | - Bruno Monier
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France.
| | - Magali Suzanne
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France.
| |
Collapse
|
3
|
Abdellatef SA, Wang H, Nakanishi J. Microtubules Disruption Alters the Cellular Structures and Mechanics Depending on Underlying Chemical Cues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2312282. [PMID: 39344221 PMCID: PMC11962689 DOI: 10.1002/smll.202312282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 09/19/2024] [Indexed: 10/01/2024]
Abstract
The extracellular matrix determines cell morphology and stiffness by manipulating the cytoskeleton. The impacts of extracellular matrix cues, including the mechanical and topographical cues on microtubules and their role in biological behaviors, are previously studied. However, there is a lack of understanding about how microtubules (MTs) are affected by environmental chemical cues, such as extracellular matrix density. Specifically, it is crucial to understand the connection between cellular morphology and mechanics induced by chemical cues and the role of microtubules in these cellular responses. To address this, surfaces with high and low cRGD (cyclic Arginine-Glycine-Aspartic acid) peptide ligand densities are used. The cRGD is diluted with a bioinert ligand to prevent surface native cellular remodeling. The cellular morphology, actin, and microtubules differ on these surfaces. Confocal fluorescence microscopes and atomic force microscopy (AFM) are used to determine the structural and mechanical cellular responses with and without microtubules. Microtubules are vital as an intracellular scaffold in elongated morphology correlated with low cRGD compared to rounded morphology in high cRGD substrates. The contributions of MTs to nucleus morphology and cellular mechanics are based on the underlying cRGD densities. Finally, this study reveals a significant correlation between MTs, actin networks, and vimentin in response to the underlying densities of cRGD.
Collapse
Affiliation(s)
- Shimaa A. Abdellatef
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
| | - Hongxin Wang
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
| | - Jun Nakanishi
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
- Graduate School of Advanced EngineeringTokyo University of Science6‐3‐1, NiijukuKatsushika‐kuTokyo125‐8585Japan
- Graduate School of Advanced Science and EngineeringWaseda University3‐4‐1 OkuboShinjuku‐kuTokyo169‐8555Japan
| |
Collapse
|
4
|
Wang H, Zhang G, Liu Y, He Y, Guo Q, Du Y, Yang C, Gao F. Glycocalyx hyaluronan removal-induced increasing of cell stiffness delays breast cancer cells progression. Cell Mol Life Sci 2025; 82:96. [PMID: 40011237 PMCID: PMC11865421 DOI: 10.1007/s00018-025-05577-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 02/28/2025]
Abstract
Triple-negative breast cancer (TNBC) cells are rich in glycocalyx (GCX) that is closely correlated with the reorganization of cytoskeletal filaments. Most studies have focused on cell membrane glycoproteins in this context, but rarely on the significance of glycosaminoglycans, particularly the hyaluronan (HA)-associated GCX. Here, we reported that removal of GCX HA could significantly increase breast cancer cells (BCCs) stiffness, leading to impaired cell growth and decreased stem-like properties. Furthermore, we found that the delay of TNBC cells progression could be restored after the cells were re-softened. Meanwhile, in vivo studies revealed that hyaluronidase (HAase)-pretreated BCCs displayed reduced tumor growth and migration. Intriguingly, we identified that ZC3H12A, a zinc-finger RNA binding protein encoded gene, was significantly upregulated after the GCX HA impairment. Of note, knockdown of ZC3H12A could soften the HAase-treated TNBC cells, implying a GCX HA-ZC3H12A regulation on cell stiffening. Taken together, our findings suggested that the breakdown of pericellular HA coat could influence TNBC cells mechanical properties which might be helpful to the future breast cancer research.
Collapse
Affiliation(s)
- Hui Wang
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Guoliang Zhang
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yiwen Liu
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yiqing He
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Qian Guo
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yan Du
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Cuixia Yang
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Feng Gao
- Department of Molecular Biology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
5
|
Nirgude S, Desai S, Ravindran F, Mhatre A, Mahadeva R, Sharma S, Rai PK, Shahana MV, Thumsi J, Choudhary B. Global transcriptome profiling of ST09 treated breast cancer cells identifies miR-197-5p/GPX3 antioxidant axis as a regulator of tumorigenesis. Int Immunopharmacol 2025; 148:114127. [PMID: 39870007 DOI: 10.1016/j.intimp.2025.114127] [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: 12/16/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/29/2025]
Abstract
ROS (Reactive Oxygen Species) has a dual role in tumorigenesis. Some cancers have high ROS conditions, and others have low ROS. TNBC thrives on high ROS compared to other Breast Cancer subtypes. Several antioxidant enzymes catalyze the detoxification of reactive oxygen species and prevent free radicals from damaging DNA and accumulation of mutation. Curcumin, a polyphenol dietary supplement, acts as a potent antioxidant, is known to reduce inflammation, and has anticancer properties. Here, we aim to understand alterations in the transcriptome (miRNA and mRNA expression) induced by ST09 in breast cancer cell lines. We identified an antioxidant system that is upregulated in breast cancer cell lines. Among the antioxidant enzymes regulated by miRNA was GPX3. A novel miRNA-mRNA antioxidant axis, miR-197-5p/GPX3, was observed in the TNBC cell line. We further validated the regulation of GPX3 by miRNA using luciferase assay. GPX3 overexpression, knockdown, and activity assay indicated the anti-tumorigenic role of GPX3 in the TNBC cell line. Further, treatment of TNBC xenograft with ST09 showed tumor reduction in vivo. ST09 potentiates the effect of standard-of-care (SOC) drug Cisplatin in vivo. ST09 can be exploited as a single chemotherapeutic agent or in combination treatment modalities, reducing the dosage of potent drugs.
Collapse
Affiliation(s)
- Snehal Nirgude
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India; Working at Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sagar Desai
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India
| | - Febina Ravindran
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India
| | - Anisha Mhatre
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India
| | - Raghunandan Mahadeva
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India
| | - Shivangi Sharma
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India; Graduate Students Registered Under Manipal Academy of Higher Education, Manipal 576104, India; Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Prashant Kumar Rai
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India; Graduate Students Registered Under Manipal Academy of Higher Education, Manipal 576104, India
| | - M V Shahana
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India; Graduate Students Registered Under Manipal Academy of Higher Education, Manipal 576104, India
| | - Jayanthi Thumsi
- BGS Gleneagles Global Hospitals, Bangalore, Karnataka, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore 560100 India.
| |
Collapse
|
6
|
Urbanska M, Ge Y, Winzi M, Abuhattum S, Ali SS, Herbig M, Kräter M, Toepfner N, Durgan J, Florey O, Dori M, Calegari F, Lolo FN, del Pozo MÁ, Taubenberger A, Cannistraci CV, Guck J. De novo identification of universal cell mechanics gene signatures. eLife 2025; 12:RP87930. [PMID: 39960760 PMCID: PMC11832173 DOI: 10.7554/elife.87930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Cell mechanical properties determine many physiological functions, such as cell fate specification, migration, or circulation through vasculature. Identifying factors that govern the mechanical properties is therefore a subject of great interest. Here, we present a mechanomics approach for establishing links between single-cell mechanical phenotype changes and the genes involved in driving them. We combine mechanical characterization of cells across a variety of mouse and human systems with machine learning-based discriminative network analysis of associated transcriptomic profiles to infer a conserved network module of five genes with putative roles in cell mechanics regulation. We validate in silico that the identified gene markers are universal, trustworthy, and specific to the mechanical phenotype across the studied mouse and human systems, and demonstrate experimentally that a selected target, CAV1, changes the mechanical phenotype of cells accordingly when silenced or overexpressed. Our data-driven approach paves the way toward engineering cell mechanical properties on demand to explore their impact on physiological and pathological cell functions.
Collapse
Affiliation(s)
- Marta Urbanska
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| | - Yan Ge
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Maria Winzi
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Shada Abuhattum
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| | - Syed Shafat Ali
- Center for Complex Network Intelligence, Tsinghua Laboratory of Brain and Intelligence, Department of Computer Science and School of Biomedical Engineering, Tsinghua UniversityBeijingChina
- Department of Computer Science and Department of Economics, Jamia Millia IslamiaNew DelhiIndia
| | - Maik Herbig
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
- Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Martin Kräter
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| | - Nicole Toepfner
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Joanne Durgan
- Signalling Programme, The Babraham InstituteCambridgeUnited Kingdom
| | - Oliver Florey
- Signalling Programme, The Babraham InstituteCambridgeUnited Kingdom
| | - Martina Dori
- Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Federico Calegari
- Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Miguel Ángel del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Anna Taubenberger
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Carlo Vittorio Cannistraci
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Center for Complex Network Intelligence, Tsinghua Laboratory of Brain and Intelligence, Department of Computer Science and School of Biomedical Engineering, Tsinghua UniversityBeijingChina
- Center for Systems Biology DresdenDresdenGermany
- Cluster of Excellence Physics of Life, Technische Universität DresdenDresdenGermany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| |
Collapse
|
7
|
Isert L, Passi M, Freystetter B, Grab M, Roidl A, Müller C, Mehta A, Sundararaghavan HG, Zahler S, Merkel OM. Cellular EMT-status governs contact guidance in an electrospun TACS-mimicking in vitro model. Mater Today Bio 2025; 30:101401. [PMID: 39759848 PMCID: PMC11699613 DOI: 10.1016/j.mtbio.2024.101401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
In this study, an advanced nanofiber breast cancer in vitro model was developed and systematically characterized including physico-chemical, cell-biological and biophysical parameters. Using electrospinning, the architecture of tumor-associated collagen signatures (TACS5 and TACS6) was mimicked. By employing a rotating cylinder or static plate collector set-up, aligned fibers (TACS5-like structures) and randomly orientated fibers (TACS6-like structures) fibers were produced, respectively. The biocompatibility of these fibers was enhanced by collagen coating, ensuring minimal toxicity and improved cell attachment. Various breast cancer cell lines (MCF7, HCC1954, MDA-MB-468, and MDA-MB-231) were cultured on these fibers to assess epithelial-to-mesenchymal transition (EMT) markers, cellular morphology, and migration. Aligned fibers (TACS5) significantly influenced EMT-related changes, promoting cellular alignment, spindle-shaped morphology and a highly migratory phenotype in mesenchymal and hybrid EMT cells (MDA-MB-468, MDA-MB-231). Conversely, epithelial cells (MCF7, HCC1954) showed limited response, but - under growth factor treatment - started to infiltrate the fibrous scaffold and underwent EMT-like changes, particularly on TACS5-mimicks, emphasizing the interplay of topographical cues and EMT induction. The biophysical analysis revealed a clear correlation between cellular EMT status and cell mechanics, with increased EMT correlating to decreased total cellular stiffness. Cancer cell mechanics, however, were found to be dynamic during biochemical and topographical EMT-induction, exceeding initial stiffness by up to 2-fold. These findings highlight the potential of TACS5-like nanofiber scaffolds in modeling the tumor microenvironment and studying cancer cell behavior and mechanics.
Collapse
Affiliation(s)
- Lorenz Isert
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| | - Mehak Passi
- Pharmaceutical Biology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benedikt Freystetter
- Department of Cardiac Surgery, Ludwig Maximilians University München, Munich, Germany
| | - Maximilian Grab
- Department of Cardiac Surgery, Ludwig Maximilians University München, Munich, Germany
| | - Andreas Roidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christoph Müller
- Center of Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Aditi Mehta
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| | | | - Stefan Zahler
- Pharmaceutical Biology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Olivia M. Merkel
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| |
Collapse
|
8
|
Bagchi A, Sarker B, Zhang J, Foston M, Pathak A. Fast yet force-effective mode of supracellular collective cell migration due to extracellular force transmission. PLoS Comput Biol 2025; 21:e1012664. [PMID: 39787053 PMCID: PMC11717197 DOI: 10.1371/journal.pcbi.1012664] [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: 02/01/2024] [Accepted: 11/21/2024] [Indexed: 01/12/2025] Open
Abstract
Cell collectives, like other motile entities, generate and use forces to move forward. Here, we ask whether environmental configurations alter this proportional force-speed relationship, since aligned extracellular matrix fibers are known to cause directed migration. We show that aligned fibers serve as active conduits for spatial propagation of cellular mechanotransduction through matrix exoskeleton, leading to efficient directed collective cell migration. Epithelial (MCF10A) cell clusters adhered to soft substrates with aligned collagen fibers (AF) migrate faster with much lesser traction forces, compared to random fibers (RF). Fiber alignment causes higher motility waves and transmission of normal stresses deeper into cell monolayer while minimizing shear stresses and increased cell-division based fluidization. By contrast, fiber randomization induces cellular jamming due to breakage in motility waves, disrupted transmission of normal stresses, and heightened shear driven flow. Using a novel motor-clutch model, we explain that such 'force-effective' fast migration phenotype occurs due to rapid stabilization of contractile forces at the migrating front, enabled by higher frictional forces arising from simultaneous compressive loading of parallel fiber-substrate connections. We also model 'haptotaxis' to show that increasing ligand connectivity (but not continuity) increases migration efficiency. According to our model, increased rate of front stabilization via higher resistance to substrate deformation is sufficient to capture 'durotaxis'. Thus, our findings reveal a new paradigm wherein the rate of leading-edge stabilization determines the efficiency of supracellular collective cell migration.
Collapse
Affiliation(s)
- Amrit Bagchi
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, Missouri, United States of America
| | - Bapi Sarker
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, Missouri, United States of America
| | - Jialiang Zhang
- Department of Energy, Environmental & Chemical Engineering, Washington University, St. Louis, Missouri, United States of America
| | - Marcus Foston
- Department of Energy, Environmental & Chemical Engineering, Washington University, St. Louis, Missouri, United States of America
| | - Amit Pathak
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, Missouri, United States of America
| |
Collapse
|
9
|
Tharp KM. Have plastic culture models prevented the discovery of effective cancer therapeutics? Br J Pharmacol 2024. [PMID: 39491545 DOI: 10.1111/bph.17387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/31/2024] [Accepted: 10/06/2024] [Indexed: 11/05/2024] Open
Abstract
Conventional cell culture techniques generally fail to recapitulate the expression profiles or functional phenotypes of the in vivo equivalents they are meant to model. These cell culture models are indispensable for preclinical drug discovery and mechanistic studies. However, if our goal is to develop effective therapies that work as intended in the human body, we must revise our cell culture models to recapitulate normal and disease physiology to ensure that we identify compounds that are useful and effective beyond our in vitro models.
Collapse
Affiliation(s)
- Kevin M Tharp
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| |
Collapse
|
10
|
Porras MÁG, Assié A, Tietjen M, Violette M, Kleiner M, Gruber-Vodicka H, Dubilier N, Leisch N. An intranuclear bacterial parasite of deep-sea mussels expresses apoptosis inhibitors acquired from its host. Nat Microbiol 2024; 9:2877-2891. [PMID: 39242818 PMCID: PMC11521996 DOI: 10.1038/s41564-024-01808-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
A limited number of bacteria are able to colonize the nuclei of eukaryotes. 'Candidatus Endonucleobacter' infects the nuclei of deep-sea mussels, where it replicates to ≥80,000 bacteria per nucleus and causes nuclei to swell to 50 times their original size. How these parasites are able to replicate and avoid apoptosis is not known. Dual RNA-sequencing transcriptomes of infected nuclei isolated using laser-capture microdissection revealed that 'Candidatus Endonucleobacter' does not obtain most of its nutrition from nuclear DNA or RNA. Instead, 'Candidatus Endonucleobacter' upregulates genes for importing and digesting sugars, lipids, amino acids and possibly mucin from its host. It likely prevents apoptosis of host cells by upregulating 7-13 inhibitors of apoptosis, proteins not previously seen in bacteria. Comparative phylogenetic analyses revealed that 'Ca. Endonucleobacter' acquired inhibitors of apoptosis through horizontal gene transfer from their hosts. Horizontal gene transfer from eukaryotes to bacteria is assumed to be rare, but may be more common than currently recognized.
Collapse
Affiliation(s)
| | - Adrien Assié
- Max Planck Institute for Marine Microbiology, Bremen, Germany
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Målin Tietjen
- Max Planck Institute for Marine Microbiology, Bremen, Germany
| | - Marlene Violette
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Manuel Kleiner
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Harald Gruber-Vodicka
- Max Planck Institute for Marine Microbiology, Bremen, Germany
- Zoological Institute, Christian-Albrechts-University, Kiel, Germany
| | - Nicole Dubilier
- Max Planck Institute for Marine Microbiology, Bremen, Germany.
| | - Nikolaus Leisch
- Max Planck Institute for Marine Microbiology, Bremen, Germany.
- European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
11
|
Khoukaz HB, Vadali M, Schoenherr A, Ramirez-Perez FI, Morales-Quinones M, Sun Z, Fujie S, Foote CA, Lyu Z, Zeng S, Augenreich MA, Cai D, Chen SY, Joshi T, Ji Y, Hill MA, Martinez-Lemus LA, Fay WP. PAI-1 Regulates the Cytoskeleton and Intrinsic Stiffness of Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2024; 44:2191-2203. [PMID: 38868940 PMCID: PMC11424258 DOI: 10.1161/atvbaha.124.320938] [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: 04/09/2024] [Accepted: 06/01/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Plasma concentration of PAI-1 (plasminogen activator inhibitor-1) correlates with arterial stiffness. Vascular smooth muscle cells (SMCs) express PAI-1, and the intrinsic stiffness of SMCs is a major determinant of total arterial stiffness. We hypothesized that PAI-1 promotes SMC stiffness by regulating the cytoskeleton and that pharmacological inhibition of PAI-1 decreases SMC and aortic stiffness. METHODS PAI-039, a specific inhibitor of PAI-1, and small interfering RNA were used to inhibit PAI-1 expression in cultured human SMCs. Effects of PAI-1 inhibition on SMC stiffness, F-actin (filamentous actin) content, and cytoskeleton-modulating enzymes were assessed. WT (wild-type) and PAI-1-deficient murine SMCs were used to determine PAI-039 specificity. RNA sequencing was performed to determine the effects of PAI-039 on SMC gene expression. In vivo effects of PAI-039 were assessed by aortic pulse wave velocity. RESULTS PAI-039 significantly reduced intrinsic stiffness of human SMCs, which was accompanied by a significant decrease in cytoplasmic F-actin content. PAI-1 gene knockdown also decreased cytoplasmic F-actin. PAI-1 inhibition significantly increased the activity of cofilin, an F-actin depolymerase, in WT murine SMCs, but not in PAI-1-deficient SMCs. RNA-sequencing analysis suggested that PAI-039 upregulates AMPK (AMP-activated protein kinase) signaling in SMCs, which was confirmed by Western blotting. Inhibition of AMPK prevented activation of cofilin by PAI-039. In mice, PAI-039 significantly decreased aortic stiffness and tunica media F-actin content without altering the elastin or collagen content. CONCLUSIONS PAI-039 decreases intrinsic SMC stiffness and cytoplasmic stress fiber content. These effects are mediated by AMPK-dependent activation of cofilin. PAI-039 also decreases aortic stiffness in vivo. These findings suggest that PAI-1 is an important regulator of the SMC cytoskeleton and that pharmacological inhibition of PAI-1 has the potential to prevent and treat cardiovascular diseases involving arterial stiffening.
Collapse
Affiliation(s)
- Hekmat B Khoukaz
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Manisha Vadali
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Alex Schoenherr
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Francisco I Ramirez-Perez
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Mariana Morales-Quinones
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Zhe Sun
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Shumpei Fujie
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan (S.F.)
| | - Christopher A Foote
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Zhen Lyu
- Electrical Engineering and Computer Science (Z.L., S.Z.), University of Missouri, Columbia
| | - Shuai Zeng
- Electrical Engineering and Computer Science (Z.L., S.Z.), University of Missouri, Columbia
| | - Marc A Augenreich
- Nutrition and Exercise Physiology (M.A.A.), University of Missouri, Columbia
| | - Dunpeng Cai
- Surgery (D.C., S.-Y.C.), University of Missouri, Columbia
| | - Shi-You Chen
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Surgery (D.C., S.-Y.C.), University of Missouri, Columbia
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (S.-Y.C., W.P.F.)
| | - Trupti Joshi
- Health Management and Informatics (T.J.), University of Missouri, Columbia
| | - Yan Ji
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Michael A Hill
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - Luis A Martinez-Lemus
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
| | - William P Fay
- Departments of Medicine (H.B.K., M.V., F.I.R.-P., M.M.-Q., Y.J., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Medical Pharmacology and Physiology (A.S., C.A.F., S.-Y.C., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Dalton Cardiovascular Research Center (Z.S., M.A.H., L.A.M.-L., W.P.F.), University of Missouri, Columbia
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (S.-Y.C., W.P.F.)
| |
Collapse
|
12
|
Jafari A, Sadeghi A, Lafouti M. Mechanical properties of human kidney cells and their effects on the atomic force microscope beam vibrations. Microsc Res Tech 2024; 87:1704-1717. [PMID: 38501545 DOI: 10.1002/jemt.24543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/20/2024]
Abstract
In the present investigation, the mechanical properties of normal and carcinomatous cells of kidney tissue (HEK-293, ACHN, respectively) were investigated using atomic force microscopy (AFM). Initially, the elastic modulus of ACHN cells was measured following chemotherapy with the anti-cancer drug Cisplatin and plasma treatment. The MTT assay was employed to ascertain the most effective dosages for incubation periods of 12, 24, 48, 72, and 96 h, guided by the IC50 concentration for cell viability during chemotherapy treatment. Analysis at these specified time points revealed a progressive increase in the elastic modulus of ACHN cells when subjected to Cisplatin-based chemotherapy. Specifically, the elastic modulus increased by 1.847, 4.416, 6.035, 8.029, and 9.727 times in comparison to untreated cells at 12, 24, 48, 72, and 96 h, respectively. ACHN cells were subsequently treated with plasma for 30 and 60 s for 24 and 48-h incubation periods. The plasma treatment increased the ACHN cell's elastic modulus. In the subsequent phase of the research, a combination of theoretical (finite element method [FEM]) and experimental methodologies was employed to investigate the resonant frequencies and magnitude of the frequency response function (FRF) concerning the movement of the AFM cantilever. This examination was conducted using ACHN cells as specimens, both before and after exposure to chemotherapy and plasma treatments. The results showed that higher sample elastic modulus increased the resonant frequency, indicating that treated cells had a higher resonant frequency than untreated cells. In conclusion, the FEM and experimental results were compared and found to be in good agreement. HIGHLIGHTS: Using Cisplatin anti-cancer drug increases the elastic modulus of ACHN cell. Applying plasma treatment increases the elastic modulus of ACHN cell. For both of the chemo and plasma therapies, increasing the incubation time increases the influence of therapies oh the cell mechanics. Using finite element modeling (FEM) the real dynamic behavior of atomic force microscope cantilever by considering human kidney cells as the soft samples is possible.
Collapse
Affiliation(s)
- Ali Jafari
- Renewable Energy Research Center, Damavand Branch, Islamic Azad University, Damavand, Iran
| | - Ali Sadeghi
- Renewable Energy Research Center, Damavand Branch, Islamic Azad University, Damavand, Iran
| | - Mansoureh Lafouti
- Renewable Energy Research Center, Damavand Branch, Islamic Azad University, Damavand, Iran
| |
Collapse
|
13
|
Qin X, Hu J, Ma S, Wu M. Estimation of multiple networks with common structures in heterogeneous subgroups. J MULTIVARIATE ANAL 2024; 202:105298. [PMID: 38433779 PMCID: PMC10907012 DOI: 10.1016/j.jmva.2024.105298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Network estimation has been a critical component of high-dimensional data analysis and can provide an understanding of the underlying complex dependence structures. Among the existing studies, Gaussian graphical models have been highly popular. However, they still have limitations due to the homogeneous distribution assumption and the fact that they are only applicable to small-scale data. For example, cancers have various levels of unknown heterogeneity, and biological networks, which include thousands of molecular components, often differ across subgroups while also sharing some commonalities. In this article, we propose a new joint estimation approach for multiple networks with unknown sample heterogeneity, by decomposing the Gaussian graphical model (GGM) into a collection of sparse regression problems. A reparameterization technique and a composite minimax concave penalty are introduced to effectively accommodate the specific and common information across the networks of multiple subgroups, making the proposed estimator significantly advancing from the existing heterogeneity network analysis based on the regularized likelihood of GGM directly and enjoying scale-invariant, tuning-insensitive, and optimization convexity properties. The proposed analysis can be effectively realized using parallel computing. The estimation and selection consistency properties are rigorously established. The proposed approach allows the theoretical studies to focus on independent network estimation only and has the significant advantage of being both theoretically and computationally applicable to large-scale data. Extensive numerical experiments with simulated data and the TCGA breast cancer data demonstrate the prominent performance of the proposed approach in both subgroup and network identifications.
Collapse
Affiliation(s)
- Xing Qin
- School of Statistics and Information, Shanghai University of International Business and Economics, Shanghai, China
| | - Jianhua Hu
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai, China
| | - Shuangge Ma
- Department of Biostatistics, Yale School of Public Health, New Haven, USA
| | - Mengyun Wu
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai, China
| |
Collapse
|
14
|
Urbanska M, Guck J. Single-Cell Mechanics: Structural Determinants and Functional Relevance. Annu Rev Biophys 2024; 53:367-395. [PMID: 38382116 DOI: 10.1146/annurev-biophys-030822-030629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
The mechanical phenotype of a cell determines its ability to deform under force and is therefore relevant to cellular functions that require changes in cell shape, such as migration or circulation through the microvasculature. On the practical level, the mechanical phenotype can be used as a global readout of the cell's functional state, a marker for disease diagnostics, or an input for tissue modeling. We focus our review on the current knowledge of structural components that contribute to the determination of the cellular mechanical properties and highlight the physiological processes in which the mechanical phenotype of the cells is of critical relevance. The ongoing efforts to understand how to efficiently measure and control the mechanical properties of cells will define the progress in the field and drive mechanical phenotyping toward clinical applications.
Collapse
Affiliation(s)
- Marta Urbanska
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
15
|
Wang HJ, Wang Y, Mirjavadi SS, Andersen T, Moldovan L, Vatankhah P, Russell B, Jin J, Zhou Z, Li Q, Cox CD, Su QP, Ju LA. Microscale geometrical modulation of PIEZO1 mediated mechanosensing through cytoskeletal redistribution. Nat Commun 2024; 15:5521. [PMID: 38951553 PMCID: PMC11217425 DOI: 10.1038/s41467-024-49833-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 06/20/2024] [Indexed: 07/03/2024] Open
Abstract
The microgeometry of the cellular microenvironment profoundly impacts cellular behaviors, yet the link between it and the ubiquitously expressed mechanosensitive ion channel PIEZO1 remains unclear. Herein, we describe a fluorescent micropipette aspiration assay that allows for simultaneous visualization of intracellular calcium dynamics and cytoskeletal architecture in real-time, under varied micropipette geometries. By integrating elastic shell finite element analysis with fluorescent lifetime imaging microscopy and employing PIEZO1-specific transgenic red blood cells and HEK cell lines, we demonstrate a direct correlation between the microscale geometry of aspiration and PIEZO1-mediated calcium signaling. We reveal that increased micropipette tip angles and physical constrictions lead to a significant reorganization of F-actin, accumulation at the aspirated cell neck, and subsequently amplify the tension stress at the dome of the cell to induce more PIEZO1's activity. Disruption of the F-actin network or inhibition of its mobility leads to a notable decline in PIEZO1 mediated calcium influx, underscoring its critical role in cellular mechanosensing amidst geometrical constraints.
Collapse
Affiliation(s)
- Haoqing Jerry Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia
| | - Yao Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Seyed Sajad Mirjavadi
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tomas Andersen
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Laura Moldovan
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia
| | - Parham Vatankhah
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Blake Russell
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Jasmine Jin
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Zijing Zhou
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St. Vincent's Clinical School, University of New South Wale, Sydney, NSW, 2010, Australia
| | - Qian Peter Su
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia.
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
16
|
Zadeh NM, Sadeghi A, Lafouti M. Mechanical Properties of Mouse Lung Cells and Their Effects on the Atomic Force Microscope Beam Vibrations. Cell Biochem Biophys 2024; 82:1079-1099. [PMID: 38713404 DOI: 10.1007/s12013-024-01259-z] [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] [Accepted: 03/19/2024] [Indexed: 05/08/2024]
Abstract
In the present investigation, the mechanical properties of mouse normal and carcinomatous (LL/2) lung tissue cells were investigated using atomic force microscopy (AFM). The normal lung cells have been derived directly from C57BL mice. Initially, the elastic modulus of LL/2 cells was measured following chemotherapy with the anti-cancer drug Cisplatin and plasma treatment. MTT evaluation was used to determine the optimal dosages for 24- and 48-h incubations based on the IC50 cell viability concentration during chemotherapy treatment. After 24 and 48 h, the results demonstrated that Cisplatin-based chemotherapy increases the elastic modulus of LL/2 cells by 1.599 and 2.308 times compared to untreated cells. LL/2 cells were subsequently treated with plasma for 30 and 60 s for 24 and 48-h incubation. The plasma treatment decreased the LL/2 cell's elastic modulus, and the time duration of plasma treatment increased the reduction amount of elastic modulus. During the second section of the study, theoretical (finite element analysis [FEM]) and experimental techniques were used to examine the resonant frequencies and magnitude of the frequency response function (FRF) of the AFM cantilever's movements when applying normal and cancerous cells before and after chemo and plasma treatments as specimens. The results indicated that increasing the samples' elastic modulus raises the resonant frequency, so the resonant frequency of treated cells as a sample is greater than untreated cells. In conclusion, the FEM and experimental results were compared and found to be in good agreement.
Collapse
Affiliation(s)
- Nazanin Maleki Zadeh
- Biomedical Department, Central Tehran Branch, Islamic Azad University, Damavand, Iran
| | - Ali Sadeghi
- Renewable Energy Research Center, Damavand Branch, Islamic Azad University, Damavand, Iran.
| | - Mansoureh Lafouti
- Renewable Energy Research Center, Damavand Branch, Islamic Azad University, Damavand, Iran
| |
Collapse
|
17
|
Kumar R, Rottner K, Rao GN. Requirement of Site-Specific Tyrosine Phosphorylation of Cortactin in Retinal Neovascularization and Vascular Leakage. Arterioscler Thromb Vasc Biol 2024; 44:366-390. [PMID: 38126170 PMCID: PMC10872470 DOI: 10.1161/atvbaha.123.320279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Retinal neovascularization is a major cause of vision impairment. Therefore, the purpose of this study is to investigate the mechanisms by which hypoxia triggers the development of abnormal and leaky blood vessels. METHODS A variety of cellular and molecular approaches as well as tissue-specific knockout mice were used to investigate the role of Cttn (cortactin) in retinal neovascularization and vascular leakage. RESULTS We found that VEGFA (vascular endothelial growth factor A) stimulates Cttn phosphorylation at Y421, Y453, and Y470 residues in human retinal microvascular endothelial cells. In addition, we observed that while blockade of Cttn phosphorylation at Y470 inhibited VEGFA-induced human retinal microvascular endothelial cell angiogenic events, suppression of Y421 phosphorylation protected endothelial barrier integrity from disruption by VEGFA. In line with these observations, while blockade of Cttn phosphorylation at Y470 negated oxygen-induced retinopathy-induced retinal neovascularization, interference with Y421 phosphorylation prevented VEGFA/oxygen-induced retinopathy-induced vascular leakage. Mechanistically, while phosphorylation at Y470 was required for its interaction with Arp2/3 and CDC6 facilitating actin polymerization and DNA synthesis, respectively, Cttn phosphorylation at Y421 leads to its dissociation from VE-cadherin, resulting in adherens junction disruption. Furthermore, whereas Cttn phosphorylation at Y470 residue was dependent on Lyn, its phosphorylation at Y421 residue required Syk activation. Accordingly, lentivirus-mediated expression of shRNA targeting Lyn or Syk levels inhibited oxygen-induced retinopathy-induced retinal neovascularization and vascular leakage, respectively. CONCLUSIONS The above observations show for the first time that phosphorylation of Cttn is involved in a site-specific manner in the regulation of retinal neovascularization and vascular leakage. In view of these findings, Cttn could be a novel target for the development of therapeutics against vascular diseases such as retinal neovascularization and vascular leakage.
Collapse
Affiliation(s)
- Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Gadiparthi N. Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
18
|
Zhang Y, Xu R, Wu J, Zhang Z, Wang Y, Yang H, Zhang S. Nanopore-related cellular death through cytoskeleton depolymerization by drug-induced ROS. Talanta 2024; 268:125355. [PMID: 37952317 DOI: 10.1016/j.talanta.2023.125355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023]
Abstract
Prostate cancer (PCa) is a malignant tumor with a very high incidence which ranks second after lung cancer. Although there are many drugs available for the treatment of PCa, their effectiveness and anti-cancer mechanisms still need to be explored. Atomic force microscopy (AFM) could characterize minor morphological changes on cell surfaces, which provides an effective method to explore the interaction between drugs and cells at the nanometer level and further investigate the mechanisms for treating PCa. In our research, AFM visualized pore-like structures in the PC3M cell membrane after treatment with the eminent anticancer agent paclitaxel (PTX). The diameter, depth and number of these pores were in a concentration and time-dependent manner. Reactive oxygen species (ROS) was shown to depolymerize the actin cytoskeleton and make the membrane more sensitive to oxidative damage, thus inducing pore information. After pretreatment with a ROS scavenger, pore formation was prevented. AFM imaging technology provides a new evaluation method for drug-targeted therapy for cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350007, China
| | - Renfeng Xu
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350007, China
| | - Jingjing Wu
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Zhenghong Zhang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350007, China
| | - Yuhuang Wang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350007, China
| | - Hongqin Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350007, China.
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
19
|
Qiu S, Qiu Y, Deng L, Nie L, Ge L, Zheng X, Jin D, Jin K, Zhou X, Su X, Cai B, Li J, Tu X, Gong L, Liu L, Liu Z, Bao Y, Ai J, Lin T, Yang L, Wei Q. Cell softness reveals tumorigenic potential via ITGB8/AKT/glycolysis signaling in a mice model of orthotopic bladder cancer. Chin Med J (Engl) 2024; 137:209-221. [PMID: 37390491 PMCID: PMC10798691 DOI: 10.1097/cm9.0000000000002710] [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: 01/29/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Bladder cancer, characterized by a high potential of tumor recurrence, has high lifelong monitoring and treatment costs. To date, tumor cells with intrinsic softness have been identified to function as cancer stem cells in several cancer types. Nonetheless, the existence of soft tumor cells in bladder tumors remains elusive. Thus, our study aimed to develop a micro-barrier microfluidic chip to efficiently isolate deformable tumor cells from distinct types of bladder cancer cells. METHODS The stiffness of bladder cancer cells was determined by atomic force microscopy (AFM). The modified microfluidic chip was utilized to separate soft cells, and the 3D Matrigel culture system was to maintain the softness of tumor cells. Expression patterns of integrin β8 (ITGB8), protein kinase B (AKT), and mammalian target of rapamycin (mTOR) were determined by Western blotting. Double immunostaining was conducted to examine the interaction between F-actin and tripartite motif containing 59 (TRIM59). The stem-cell-like characteristics of soft cells were explored by colony formation assay and in vivo studies upon xenografted tumor models. RESULTS Using our newly designed microfluidic approach, we identified a small fraction of soft tumor cells in bladder cancer cells. More importantly, the existence of soft tumor cells was confirmed in clinical human bladder cancer specimens, in which the number of soft tumor cells was associated with tumor relapse. Furthermore, we demonstrated that the biomechanical stimuli arising from 3D Matrigel activated the F-actin/ITGB8/TRIM59/AKT/mTOR/glycolysis pathways to enhance the softness and tumorigenic capacity of tumor cells. Simultaneously, we detected a remarkable up-regulation in ITGB8, TRIM59, and phospho-AKT in clinical bladder recurrent tumors compared with their non-recurrent counterparts. CONCLUSIONS The ITGB8/TRIM59/AKT/mTOR/glycolysis axis plays a crucial role in modulating tumor softness and stemness. Meanwhile, the soft tumor cells become more sensitive to chemotherapy after stiffening, that offers new insights for hampering tumor progression and recurrence.
Collapse
Affiliation(s)
- Shi Qiu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Department of Molecular Oncology, Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Bellinzona 6500, Switzerland
| | - Yaqi Qiu
- Department of Science and Drug Technology, University of Turin, Turin, Italy
| | - Linghui Deng
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liming Ge
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610000, China
| | - Xiaonan Zheng
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Di Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianghong Zhou
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyang Su
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Boyu Cai
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiakun Li
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiang Tu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Lina Gong
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yige Bao
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianhai Lin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
20
|
Ozdil B, Calik-Kocaturk D, Altunayar-Unsalan C, Acikgoz E, Oltulu F, Gorgulu V, Uysal A, Oktem G, Unsalan O, Guler G, Aktug H. Differences and similarities in biophysical and biological characteristics between U87 MG glioblastoma and astrocyte cells. Histochem Cell Biol 2024; 161:43-57. [PMID: 37700206 DOI: 10.1007/s00418-023-02234-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2023] [Indexed: 09/14/2023]
Abstract
Current cancer studies focus on molecular-targeting diagnostics and interactions with surroundings; however, there are still gaps in characterization based on topological differences and elemental composition. Glioblastoma (GBM cells; GBMCs) is an astrocytic aggressive brain tumor. At the molecular level, GBMCs and astrocytes may differ, and cell elemental/topological analysis is critical for identifying potential new cancer targets. Here, we used U87 MG cells for GBMCS. U87 MG cell lines, which are frequently used in glioblastoma research, are an important tool for studying the various features and underlying mechanisms of this aggressive brain tumor. For the first time, atomic force microscopy (AFM), scanning electron microscopy (SEM) accompanied by energy-dispersive X-ray spectroscopy (EDS), and X-ray photoelectron spectroscopy (XPS) are used to report the topology and chemistry of cancer (U87 MG) and healthy (SVG p12) cells. In addition, F-actin staining and cytoskeleton-based gene expression analyses were performed. The degree of gene expression for genes related to the cytoskeleton was similar; however, the intensity of F-actin, anisotropy values, and invasion-related genes were different. Morphologically, GBMCs were longer and narrower while astrocytes were shorter and more disseminated based on AFM. Furthermore, the roughness values of these cells differed slightly between the two call types. In contrast to the rougher astrocyte surfaces in the lamellipodial area, SEM-EDS analysis showed that elongated GBMCs displayed filopodial protrusions. Our investigation provides considerable further insight into rapid cancer cell characterization in terms of a combinatorial spectroscopic and microscopic approach.
Collapse
Affiliation(s)
- Berrin Ozdil
- Department of Histology and Embryology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey
- Department of Histology and Embryology, Faculty of Medicine, Suleyman Demirel University, 32260, Isparta, Turkey
| | | | - Cisem Altunayar-Unsalan
- Central Research Testing and Analysis Laboratory Research and Application Center, Ege University, 35100, Bornova, Izmir, Turkey.
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yüzüncü Yıl University, 65080, Van, Turkey
| | - Fatih Oltulu
- Department of Histology and Embryology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey.
| | - Volkan Gorgulu
- Department of Histology and Embryology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey
| | - Aysegul Uysal
- Department of Histology and Embryology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey
| | - Gulperi Oktem
- Department of Histology and Embryology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey
| | - Ozan Unsalan
- Department of Physics, Faculty of Science, Ege University, 35100, Izmir, Turkey
| | - Gunnur Guler
- Department of Physics, Biophysics Laboratory, Izmir Institute of Technology, 35430, Izmir, Turkey
| | - Huseyin Aktug
- Department of Histology and Embryology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey
| |
Collapse
|
21
|
Arce FT, Younger S, Gaber AA, Mascarenhas JB, Rodriguez M, Dudek SM, Garcia JGN. Lamellipodia dynamics and microrheology in endothelial cell paracellular gap closure. Biophys J 2023; 122:4730-4747. [PMID: 37978804 PMCID: PMC10754712 DOI: 10.1016/j.bpj.2023.11.016] [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: 01/15/2023] [Revised: 05/06/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023] Open
Abstract
Vascular endothelial cells (ECs) form a semipermeable barrier separating vascular contents from the interstitium, thereby regulating the movement of water and molecular solutes across small intercellular gaps, which are continuously forming and closing. Under inflammatory conditions, however, larger EC gaps form resulting in increased vascular leakiness to circulating fluid, proteins, and cells, which results in organ edema and dysfunction responsible for key pathophysiologic findings in numerous inflammatory disorders. In this study, we extend our earlier work examining the biophysical properties of EC gap formation and now address the role of lamellipodia, thin sheet-like membrane projections from the leading edge, in modulating EC spatial-specific contractile properties and gap closure. Micropillars, fabricated by soft lithography, were utilized to form reproducible paracellular gaps in human lung ECs. Using time-lapse imaging via optical microscopy, rates of EC gap closure and motility were measured with and without EC stimulation with the barrier-enhancing sphingolipid, sphingosine-1-phosphate. Peripheral ruffle formation was ubiquitous during gap closure. Kymographs were generated to quantitatively compare the lamellipodia dynamics of sphingosine-1-phosphate-stimulated and -unstimulated ECs. Utilizing atomic force microscopy, we characterized the viscoelastic behavior of EC lamellipodia. Our results indicate decreased stiffness and increased liquid-like behavior of expanding lamellipodia compared with regions away from the cellular edge (lamella and cell body) during EC gap closure, results in sync with the rapid kinetics of protrusion/retraction motion. We hypothesize this dissipative EC behavior during gap closure is linked to actomyosin cytoskeletal rearrangement and decreased cross-linking during lamellipodia expansion. In summary, these studies of the kinetic and mechanical properties of EC lamellipodia and ruffles at gap boundaries yield insights into the mechanisms of vascular barrier restoration and potentially a model system for examining the druggability of lamellipodial protein targets to enhance vascular barrier integrity.
Collapse
Affiliation(s)
- Fernando Teran Arce
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida.
| | - Scott Younger
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Amir A Gaber
- Department of Medicine, University of Arizona, Tucson, Arizona
| | | | - Marisela Rodriguez
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida; Department of Medicine, University of Arizona, Tucson, Arizona
| | - Steven M Dudek
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois
| | - Joe G N Garcia
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida.
| |
Collapse
|
22
|
Wang Y, Wang J, Li Q, Xuan R, Guo Y, He P, Chao T. Characterization of MicroRNA expression profiles in the ovarian tissue of goats during the sexual maturity period. J Ovarian Res 2023; 16:234. [PMID: 38062510 PMCID: PMC10704810 DOI: 10.1186/s13048-023-01318-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The ovary is an important reproductive organ in mammals, and its development directly affects the sexual maturity and reproductive capacity of individuals. MicroRNAs (miRNAs) are recognized as regulators of reproductive physiological processes in various animals and have been shown to regulate ovarian development through typical targeting and translational repression. However, little is known about the regulatory role of miRNAs in ovarian tissue development during sexual maturity in goats. To comprehensively profile the different physiological stages of sexual maturation in goats, we performed small-RNA sequencing of ovarian tissue samples collected at four specific time points (1 day after birth (D1), 2 months old (M2), 4 months old (M4), and 6 months old (M6)). In addition, we used ELISAs to measure serum levels of reproductive hormones to study their temporal changes. RESULTS The results showed that serum levels of gonadotropin-releasing hormone, follicle-stimulating hormone, luteinizing hormone, oestradiol, progesterone, oxytocin, and prolactin were lower in goats at the D1 stage than at the other three developmental stages (P < 0.05). The secretion patterns of these seven hormones show a similar trend, with hormone levels reaching their peaks at 4 months of age. A total of 667 miRNAs were detected in 20 libraries, and 254 differentially expressed miRNAs and 3 groups of miRNA clusters that had unique expression patterns were identified (|log2-fold change|> 1, FDR < 0.05) in the 6 comparison groups. RT‒qPCR was employed to confirm that the expression pattern of the 15 selected miRNAs was consistent with the Illumina sequencing results. Gene ontology analyses revealed significant enrichment of GO terms such as cell proliferation regulation, epithelial cell development, and amino acid transport, as well as important signaling pathways including the MAPK signaling pathway, the PI3K-Akt signaling pathway, and the oestrogen signaling pathway. Further miRNA‒mRNA regulation network analysis revealed that 8 differentially expressed miRNAs (chi-miR-1343, chi-miR-328-3p, chi-miR-877-3p, chi-miR-296-3p, chi-miR-128-5p, chi-miR-331-3p, chi-miR-342-5p and chi-miR-34a) have important regulatory roles in ovarian cell proliferation, hormone secretion and metabolism-related biological processes. CONCLUSIONS Overall, our study investigated the changes in serum hormone and miRNA levels in the ovaries. These data provide a valuable resource for understanding the molecular regulatory mechanisms of miRNAs in ovarian tissue during the sexual maturity period in goats.
Collapse
Affiliation(s)
- Yanyan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Qing Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Rong Xuan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Yanfei Guo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Peipei He
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China.
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, China.
| |
Collapse
|
23
|
Crozet F, Levayer R. Emerging roles and mechanisms of ERK pathway mechanosensing. Cell Mol Life Sci 2023; 80:355. [PMID: 37947896 PMCID: PMC10638131 DOI: 10.1007/s00018-023-05007-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
The coupling between mechanical forces and modulation of cell signalling pathways is essential for tissue plasticity and their adaptation to changing environments. Whilst the number of physiological and pathological relevant roles of mechanotransduction has been rapidly expanding over the last decade, studies have been mostly focussing on a limited number of mechanosensitive pathways, which include for instance Hippo/YAP/TAZ pathway, Wnt/β-catenin or the stretch-activated channel Piezo. However, the recent development and spreading of new live sensors has provided new insights into the contribution of ERK pathway in mechanosensing in various systems, which emerges now as a fast and modular mechanosensitive pathway. In this review, we will document key in vivo and in vitro examples that have established a clear link between cell deformation, mechanical stress and modulation of ERK signalling, comparing the relevant timescale and mechanical stress. We will then discuss different molecular mechanisms that have been proposed so far, focussing on the epistatic link between mechanics and ERK and discussing the relevant cellular parameters affecting ERK signalling. We will finish by discussing the physiological and the pathological consequences of the link between ERK and mechanics, outlining how this interplay is instrumental for self-organisation and long-range cell-cell coordination.
Collapse
Affiliation(s)
- Flora Crozet
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 Rue du Dr. Roux, 75015, Paris, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 Rue du Dr. Roux, 75015, Paris, France.
| |
Collapse
|
24
|
Woodcock E, Gorelkin PV, Goff PS, Edwards CRW, Zhang Y, Korchev Y, Sviderskaya EV. Measuring Melanoma Nanomechanical Properties in Relation to Metastatic Ability and Anti-Cancer Drug Treatment Using Scanning Ion Conductance Microscopy. Cells 2023; 12:2401. [PMID: 37830615 PMCID: PMC10571876 DOI: 10.3390/cells12192401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
A cell's mechanical properties have been linked to cancer development, motility and metastasis and are therefore an attractive target as a universal, reliable cancer marker. For example, it has been widely published that cancer cells show a lower Young's modulus than their non-cancerous counterparts. Furthermore, the effect of anti-cancer drugs on cellular mechanics may offer a new insight into secondary mechanisms of action and drug efficiency. Scanning ion conductance microscopy (SICM) offers a nanoscale resolution, non-contact method of nanomechanical data acquisition. In this study, we used SICM to measure the nanomechanical properties of melanoma cell lines from different stages with increasing metastatic ability. Young's modulus changes following treatment with the anti-cancer drugs paclitaxel, cisplatin and dacarbazine were also measured, offering a novel perspective through the use of continuous scan mode SICM. We found that Young's modulus was inversely correlated to metastatic ability in melanoma cell lines from radial growth, vertical growth and metastatic phases. However, Young's modulus was found to be highly variable between cells and cell lines. For example, the highly metastatic cell line A375M was found to have a significantly higher Young's modulus, and this was attributed to a higher level of F-actin. Furthermore, our data following nanomechanical changes after 24 hour anti-cancer drug treatment showed that paclitaxel and cisplatin treatment significantly increased Young's modulus, attributed to an increase in microtubules. Treatment with dacarbazine saw a decrease in Young's modulus with a significantly lower F-actin corrected total cell fluorescence. Our data offer a new perspective on nanomechanical changes following drug treatment, which may be an overlooked effect. This work also highlights variations in cell nanomechanical properties between previous studies, cancer cell lines and cancer types and questions the usefulness of using nanomechanics as a diagnostic or prognostic tool.
Collapse
Affiliation(s)
- Emily Woodcock
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London SW17 0RE, UK; (E.W.)
- Department of Medicine, Imperial College London, W12 0NN London, UK (Y.K.)
| | - Peter V. Gorelkin
- Research Laboratory of Biophysics, National University of Science and Technology MISiS, Moscow 119049, Russia
| | - Philip S. Goff
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London SW17 0RE, UK; (E.W.)
| | | | - Yanjun Zhang
- Department of Medicine, Imperial College London, W12 0NN London, UK (Y.K.)
- Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yuri Korchev
- Department of Medicine, Imperial College London, W12 0NN London, UK (Y.K.)
- Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Elena V. Sviderskaya
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London SW17 0RE, UK; (E.W.)
| |
Collapse
|
25
|
Cho Y, Kim J, Park J, Doh J. Surface nanotopography and cell shape modulate tumor cell susceptibility to NK cell cytotoxicity. MATERIALS HORIZONS 2023; 10:4532-4540. [PMID: 37559559 DOI: 10.1039/d3mh00367a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Natural killer (NK) cells are innate cytotoxic lymphocytes exerting cytotoxicity against virally infected cells and tumor cells. NK cell cytotoxicity is primarily determined by biochemical signals received from ligands expressed on target cell surfaces, but it is also possible that biophysical environments of tumor cells, such as nanoscale surface topography typically existing on extracellular matrixes (ECMs) or cell morphology determined by ECM spaces or cell density, regulate NK cell cytotoxicity. In this study, micro/nanofabrication technology was applied to examine this possibility. Tumor cells were plated on flat or nanogrooved surfaces, or micropatterned into circular or elliptical geometries, and the effects of surface topography and tumor cell morphology on NK cell cytotoxicity were investigated. NK cells exhibited significantly higher cytotoxicity against tumor cells on nanogrooved surfaces or tumor cells in elliptical patterns than tumor cells on flat surfaces or tumor cells in circular patterns, respectively. The amounts of stress fiber formation in tumor cells positively correlated with NK cell cytotoxicity, indicating that increased cellular tension of tumor cells, either mediated by nanogrooved surfaces or elongated morphologies, was a key factor regulating NK cell cytotoxicity. These results may provide insight into the design of NK cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Yongbum Cho
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology, 77, Cheongam-ro, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - JangHyuk Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul, South Korea.
| | - Jeehun Park
- SOFT Foundry Institute, Seoul National University, Seoul, South Korea.
| | - Junsang Doh
- Department of Materials Science and Engineering, Seoul National University, Seoul, South Korea.
- SOFT Foundry Institute, Seoul National University, Seoul, South Korea.
- Institute of Engineering Research, BioMAX, Seoul National University, Seoul, South Korea
| |
Collapse
|
26
|
Jablonski KP, Beerenwinkel N. Coherent pathway enrichment estimation by modeling inter-pathway dependencies using regularized regression. Bioinformatics 2023; 39:btad522. [PMID: 37610338 PMCID: PMC10471899 DOI: 10.1093/bioinformatics/btad522] [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: 11/12/2022] [Revised: 07/04/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023] Open
Abstract
MOTIVATION Gene set enrichment methods are a common tool to improve the interpretability of gene lists as obtained, for example, from differential gene expression analyses. They are based on computing whether dysregulated genes are located in certain biological pathways more often than expected by chance. Gene set enrichment tools rely on pre-existing pathway databases such as KEGG, Reactome, or the Gene Ontology. These databases are increasing in size and in the number of redundancies between pathways, which complicates the statistical enrichment computation. RESULTS We address this problem and develop a novel gene set enrichment method, called pareg, which is based on a regularized generalized linear model and directly incorporates dependencies between gene sets related to certain biological functions, for example, due to shared genes, in the enrichment computation. We show that pareg is more robust to noise than competing methods. Additionally, we demonstrate the ability of our method to recover known pathways as well as to suggest novel treatment targets in an exploratory analysis using breast cancer samples from TCGA. AVAILABILITY AND IMPLEMENTATION pareg is freely available as an R package on Bioconductor (https://bioconductor.org/packages/release/bioc/html/pareg.html) as well as on https://github.com/cbg-ethz/pareg. The GitHub repository also contains the Snakemake workflows needed to reproduce all results presented here.
Collapse
Affiliation(s)
- Kim Philipp Jablonski
- Department of Biosystems Science and Engineering, ETH Zurich, Basel 4058, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel 4058, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel 4058, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel 4058, Switzerland
| |
Collapse
|
27
|
Fu X, Xu M, Yu Z, Gu W, Zhang Z, Zhang B, Wang X, Su Z, Zhang C. Staphylococcal Enterotoxin C2 Mutant-Induced Antitumor Immune Response Is Controlled by CDC42/MLC2-Mediated Tumor Cell Stiffness. Int J Mol Sci 2023; 24:11796. [PMID: 37511553 PMCID: PMC10380429 DOI: 10.3390/ijms241411796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/03/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
As a biological macromolecule, the superantigen staphylococcal enterotoxin C2 (SEC2) is one of the most potent known T-cell activators, and it induces massive cytotoxic granule production. With this property, SEC2 and its mutants are widely regarded as immunomodulating agents for cancer therapy. In a previous study, we constructed an MHC-II-independent mutant of SEC2, named ST-4, which exhibits enhanced immunocyte stimulation and antitumor activity. However, tumor cells have different degrees of sensitivity to SEC2/ST-4. The mechanisms of immune resistance to SEs in cancer cells have not been investigated. Herein, we show that ST-4 could activate more powerful human lymphocyte granule-based cytotoxicity than SEC2. The results of RNA-seq and atomic force microscopy (AFM) analysis showed that, compared with SKOV3 cells, the softer ES-2 cells could escape from SEC2/ST-4-induced cytotoxic T-cell-mediated apoptosis by regulating cell softness through the CDC42/MLC2 pathway. Conversely, after enhancing the stiffness of cancer cells by a nonmuscle myosin-II-specific inhibitor, SEC2/ST-4 exhibited a significant antitumor effect against ES-2 cells by promoting perforin-dependent apoptosis and the S-phase arrest. Taken together, these data suggest that cell stiffness could be a key factor of resistance to SEs in ovarian cancer, and our findings may provide new insight for SE-based tumor immunotherapy.
Collapse
Affiliation(s)
- Xuanhe Fu
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- Department of Immunology, Shenyang Medical College, No. 146 Huanghe North Street, Shenyang 110034, China
- Key Laboratory of Superantigen Research of Liao Ning Province, Shenyang 110016, China
| | - Mingkai Xu
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- Key Laboratory of Superantigen Research of Liao Ning Province, Shenyang 110016, China
| | - Zhixiong Yu
- Department of Immunology, Shenyang Medical College, No. 146 Huanghe North Street, Shenyang 110034, China
| | - Wu Gu
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhichun Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bowen Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiujuan Wang
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- Key Laboratory of Superantigen Research of Liao Ning Province, Shenyang 110016, China
| | - Zhencheng Su
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- Key Laboratory of Superantigen Research of Liao Ning Province, Shenyang 110016, China
| | - Chenggang Zhang
- Institute of Applied Ecology, Chinese Academy of Sciences, No. 72 Wenhua Road, Shenyang 110016, China
- Key Laboratory of Superantigen Research of Liao Ning Province, Shenyang 110016, China
| |
Collapse
|
28
|
Brito C, Pereira JM, Mesquita FS, Cabanes D, Sousa S. Src-Dependent NM2A Tyrosine Phosphorylation Regulates Actomyosin Remodeling. Cells 2023; 12:1871. [PMID: 37508535 PMCID: PMC10377941 DOI: 10.3390/cells12141871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Non-muscle myosin 2A (NM2A) is a key cytoskeletal enzyme that, along with actin, assembles into actomyosin filaments inside cells. NM2A is fundamental for cell adhesion and motility, playing important functions in different stages of development and during the progression of viral and bacterial infections. Phosphorylation events regulate the activity and the cellular localization of NM2A. We previously identified the tyrosine phosphorylation of residue 158 (pTyr158) in the motor domain of the NM2A heavy chain. This phosphorylation can be promoted by Listeria monocytogenes infection of epithelial cells and is dependent on Src kinase; however, its molecular role is unknown. Here, we show that the status of pTyr158 defines cytoskeletal organization, affects the assembly/disassembly of focal adhesions, and interferes with cell migration. Cells overexpressing a non-phosphorylatable NM2A variant or expressing reduced levels of Src kinase display increased stress fibers and larger focal adhesions, suggesting an altered contraction status consistent with the increased NM2A activity that we also observed. We propose NM2A pTyr158 as a novel layer of regulation of actomyosin cytoskeleton organization.
Collapse
Affiliation(s)
- Cláudia Brito
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
- MCBiology PhD Program-Instituto de Ciências Biomédicas Abel Salazar-ICBAS, University of Porto, 4050-313 Porto, Portugal
| | - Joana M Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
- MCBiology PhD Program-Instituto de Ciências Biomédicas Abel Salazar-ICBAS, University of Porto, 4050-313 Porto, Portugal
| | - Francisco S Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| | - Didier Cabanes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| |
Collapse
|
29
|
Millán M, Villarreal L, D'Aiuto N, Bologna-Molina R, Sotelo-Silveira J, Benech JC, Hochmann J, Arocena M. Mechanical profile of human keratinocytes expressing HPV-18 oncogenes. Biochem Biophys Res Commun 2023; 657:86-91. [PMID: 36996545 DOI: 10.1016/j.bbrc.2023.03.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
During tumorigenesis, the mechanical properties of cancer cells change markedly, with decreased stiffness often accompanying a more invasive phenotype. Less is known about the changes in mechanical parameters at intermediate stages in the process of malignant transformation. We have recently developed a pre-tumoral cell model by stably transducing the immortalized but non-tumorigenic human keratinocyte cell line HaCaT with the E5, E6 and E7 oncogenes from HPV-18, one of the leading causes of cervical cancer and other types of cancer worldwide. We have used atomic force microscopy (AFM) to measure cell stiffness and to obtain mechanical maps of parental HaCaT and HaCaT E5/E6/E7-18 cell lines. We observed a significant decrease in Young's modulus in HaCaT E5/E6/E7-18 cells measured by nanoindentation in the central region, as well as decreased cell rigidity in regions of cell-cell contact measured by Peakforce Quantitative Nanomechanical Mapping (PF-QNM). As a morphological correlate, HaCaT E5/E6/E7-18 cells displayed a significantly rounder cell shape than parental HaCaT cells. Our results therefore show that decreased stiffness with concomitant perturbations in cell shape are early mechanical and morphological changes during the process of malignant transformation.
Collapse
Affiliation(s)
- Magdalena Millán
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay
| | - Lihuén Villarreal
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay
| | - Natali D'Aiuto
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Cátedra de Bioquímica y Biofísica, Facultad de Odontología, Universidad de la República, Uruguay
| | - Ronell Bologna-Molina
- Departamento de Patología Molecular, Facultad de Odontología, Universidad de la República, Uruguay
| | - José Sotelo-Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Uruguay
| | - Juan C Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay
| | - Jimena Hochmann
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Uruguay.
| | - Miguel Arocena
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Cátedra de Bioquímica y Biofísica, Facultad de Odontología, Universidad de la República, Uruguay.
| |
Collapse
|
30
|
Song Z, Cheng Y, Chen M, Xie X. Macrophage polarization in bone implant repair: A review. Tissue Cell 2023; 82:102112. [PMID: 37257287 DOI: 10.1016/j.tice.2023.102112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/10/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
Macrophages (MΦ) are highly adaptable and functionally polarized cells that play a crucial role in various physiological and pathological processes. Typically, MΦ differentiate into two distinct subsets: the proinflammatory (M1) and anti-inflammatory (M2) phenotypes. Due to their potent immunomodulatory and anti-inflammatory properties, MΦ have garnered significant attention in recent decades. In the context of bone implant repair, the immunomodulatory function of MΦ is of paramount importance. Depending on their polarization phenotype, MΦ can exert varying effects on osteogenesis, angiogenesis, and the inflammatory response around the implant. This paper provides an overview of the immunomodulatory and inflammatory effects of MΦ polarization in the repair of bone implants.
Collapse
Affiliation(s)
- Zhengzheng Song
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China; Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China
| | - Yuxi Cheng
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China; Xiangya School of Stomatology, Central South University, Changsha 410008, Hunan, China
| | - Minmin Chen
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China.
| | - Xiaoli Xie
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha 410078, Hunan, China; Hunan Key Laboratory of Oral Health Research, Changsha 410008, Hunan, China.
| |
Collapse
|
31
|
Yan R, Liu D, Wang J, Liu M, Guo H, Bai J, Yang S, Chang J, Yao Z, Yang Z, Blom T, Zhou K. miR-137-LAPTM4B regulates cytoskeleton organization and cancer metastasis via the RhoA-LIMK-Cofilin pathway in osteosarcoma. Oncogenesis 2023; 12:25. [PMID: 37147294 PMCID: PMC10163001 DOI: 10.1038/s41389-023-00471-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/07/2023] Open
Abstract
Osteosarcoma (OS) is a rare malignant bone tumor but is one leading cause of cancer mortality in childhood and adolescence. Cancer metastasis accounts for the primary reason for treatment failure in OS patients. The dynamic organization of the cytoskeleton is fundamental for cell motility, migration, and cancer metastasis. Lysosome Associated Protein Transmembrane 4B (LAPTM4B) is an oncogene participating in various biological progress central to cancer biogenesis. However, the potential roles of LAPTM4B in OS and the related mechanisms remain unknown. Here, we established the elevated LAPTM4B expression in OS, and it is essential in regulating stress fiber organization through RhoA-LIMK-cofilin signaling pathway. In terms of mechanism, our data revealed that LAPTM4B promotes RhoA protein stability by suppressing the ubiquitin-mediated proteasome degradation pathway. Moreover, our data show that miR-137, rather than gene copy number and methylation status, contributes to the upregulation of LAPTM4B in OS. We report that miR-137 is capable of regulating stress fiber arrangement, OS cell migration, and metastasis via targeting LAPTM4B. Combining results from cells, patients' tissue samples, the animal model, and cancer databases, this study further suggests that the miR-137-LAPTM4B axis represents a clinically relevant pathway in OS progression and a viable target for novel therapeutics.
Collapse
Affiliation(s)
- Ruyu Yan
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Dan Liu
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Junjie Wang
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Minxia Liu
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, 00290, Finland
| | - Hongjuan Guo
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jing Bai
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shuo Yang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Jun Chang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhihong Yao
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Tomas Blom
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, 00014, Finland.
| | - Kecheng Zhou
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, 00014, Finland.
| |
Collapse
|
32
|
Lima LTFD, Ganzella FADO, Cardoso GC, Pires VDS, Chequin A, Santos GL, Braun-Prado K, Galindo CM, Braz Junior O, Molento MB, Acco A, Adami ER, Costa ET, Cavichiolo Franco CR, Klassen G, Ramos EADS. l-carvone decreases breast cancer cells adhesion, migration, and invasion by suppressing FAK activation. Chem Biol Interact 2023; 378:110480. [PMID: 37059214 DOI: 10.1016/j.cbi.2023.110480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/24/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Breast cancer is one of the most common types of cancer in the world and current therapeutic strategies present severe drawbacks. l-carvone (CRV), a monoterpene found in Mentha spicata (spearmint), has been reported to have potent anti-inflammatory activity. Here, we examined the role of CRV in breast cancer cell adhesion, migration and invasion in vitro and how this component could suppress the growth of Ehrlich carcinoma-bearing mice. In vivo, treatment with CRV significantly decreased tumor growth, increased tumor necrosis area, and reduced the expression of VEGF and HIF-1α in Ehrlich carcinoma-bearing mice. Furthermore, the anticancer efficacy of CRV was similar to currently used chemotherapy (Methotrexate), and the combination of CRV with MTX potentiated the chemotherapy effects. Further mechanistic investigation in vitro revealed that CRV modulates the interaction of breast cancer cells with the extracellular matrix (ECM) by disrupting focal adhesion, which was shown by scanning electron microscopy (SEM) and immunofluorescence. Moreover, CRV caused a decrease in β1-integrin expression and inhibited focal adhesion kinase (FAK) activation. FAK is one of the most important downstream activators of several metastatic processes, including MMP-2 mediated invasion and HIF-1α/VEGF angiogenesis stimulus, both of which were found to be reduced in MDA-MB-231 cells exposed to CRV. Our results provide new insight about targeting β1-integrin/FAK signaling pathway with CRV, which could be a new potential agent in the treatment of breast cancer.
Collapse
Affiliation(s)
- Lucas Trevisan França de Lima
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Gabriela Casani Cardoso
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Verônica Dos Santos Pires
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Andressa Chequin
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Giulia Luiza Santos
- Molecular Oncology Center, Research and Education Institute, Hospital Sirio-Libanes, São Paulo, SP, Brazil
| | - Karin Braun-Prado
- Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Odair Braz Junior
- Pos-graduate Program of Cellular and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo Beltrão Molento
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Veterinary Medicine, Federal University of Parana, Curitiba, PR, Brazil
| | - Alexandra Acco
- Pos-graduate Program of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Eliana Rezende Adami
- Pos-graduate Program of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Erico Tosoni Costa
- Molecular Oncology Center, Research and Education Institute, Hospital Sirio-Libanes, São Paulo, SP, Brazil
| | | | - Giseli Klassen
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Edneia Amancio de Souza Ramos
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil.
| |
Collapse
|
33
|
Beckmann A, Ramirez P, Gamez M, Gonzalez E, De Mange J, Bieniek KF, Ray WJ, Frost B. Moesin is an effector of tau-induced actin overstabilization, cell cycle activation, and neurotoxicity in Alzheimer's disease. iScience 2023; 26:106152. [PMID: 36879821 PMCID: PMC9984563 DOI: 10.1016/j.isci.2023.106152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/01/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
In Alzheimer's disease, neurons acquire phenotypes that are also present in various cancers, including aberrant activation of the cell cycle. Unlike cancer, cell cycle activation in post-mitotic neurons is sufficient to induce cell death. Multiple lines of evidence suggest that abortive cell cycle activation is a consequence of pathogenic forms of tau, a protein that drives neurodegeneration in Alzheimer's disease and related "tauopathies." Here we combine network analyses of human Alzheimer's disease and mouse models of Alzheimer's disease and primary tauopathy with studies in Drosophila to discover that pathogenic forms of tau drive cell cycle activation by disrupting a cellular program involved in cancer and the epithelial-mesenchymal transition (EMT). Moesin, an EMT driver, is elevated in cells harboring disease-associated phosphotau, over-stabilized actin, and ectopic cell cycle activation. We further find that genetic manipulation of Moesin mediates tau-induced neurodegeneration. Taken together, our study identifies novel parallels between tauopathy and cancer.
Collapse
Affiliation(s)
- Adrian Beckmann
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Paulino Ramirez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Maria Gamez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Elias Gonzalez
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jasmine De Mange
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - Kevin F. Bieniek
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| | - William J. Ray
- The Neurodegeneration Consortium, Therapeutics Discovery Division, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bess Frost
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, San Antonio, TX, USA
- University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
34
|
Lazzarini R, Eléxpuru-Zabaleta M, Piva F, Giulietti M, Fulgenzi G, Tartaglione MF, Zingaretti L, Tagliabracci A, Valentino M, Santarelli L, Bracci M. Effects of extremely low-frequency magnetic fields on human MDA-MB-231 breast cancer cells: proteomic characterization. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114650. [PMID: 36805133 DOI: 10.1016/j.ecoenv.2023.114650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 01/29/2023] [Accepted: 02/11/2023] [Indexed: 06/18/2023]
Abstract
Extremely low-frequency electromagnetic fields (ELF-MF) can modify the cell viability and regulatory processes of some cell types, including breast cancer cells. Breast cancer is a multifactorial disease where a role for ELF-MF cannot be excluded. ELF-MF may influence the biological properties of breast cells through molecular mechanisms and signaling pathways that are still unclear. This study analyzed the changes in the cell viability, cellular morphology, oxidative stress response and alteration of proteomic profile in breast cancer cells (MDA-MB-231) exposed to ELF-MF (50 Hz, 1 mT for 4 h). Non-tumorigenic human breast cells (MCF-10A) were used as control cells. Exposed MDA-MB-231 breast cancer cells increased their viability and live cell number and showed a higher density and length of filopodia compared with the unexposed cells. In addition, ELF-MF induced an increase of the mitochondrial ROS levels and an alteration of mitochondrial morphology. Proteomic data analysis showed that ELF-MF altered the expression of 328 proteins in MDA-MB-231 cells and of 242 proteins in MCF-10A cells. Gene Ontology term enrichment analysis demonstrated that in both cell lines ELF-MF exposure up-regulated the genes enriched in "focal adhesion" and "mitochondrion". The ELF-MF exposure decreased the adhesive properties of MDA-MB-231 cells and increased the migration and invasion cell abilities. At the same time, proteomic analysis, confirmed by Real Time PCR, revealed that transcription factors associated with cellular reprogramming were upregulated in MDA-MB-231 cells and downregulated in MCF-10A cells after ELF-MF exposure. MDA-MB-231 breast cancer cells exposed to 1 mT 50 Hz ELF-MF showed modifications in proteomic profile together with changes in cell viability, cellular morphology, oxidative stress response, adhesion, migration and invasion cell abilities. The main signaling pathways involved were relative to focal adhesion, mitochondrion and cellular reprogramming.
Collapse
Affiliation(s)
- Raffaella Lazzarini
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy.
| | - Maria Eléxpuru-Zabaleta
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain.
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy.
| | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy.
| | - Gianluca Fulgenzi
- Experimental Pathology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy.
| | - Maria Fiorella Tartaglione
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy.
| | - Laura Zingaretti
- Occupational Medicine Unit, Marche University Hospital, 60126 Ancona, Italy.
| | - Adriano Tagliabracci
- Department of Excellence of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy.
| | - Matteo Valentino
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy.
| | - Lory Santarelli
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy.
| | - Massimo Bracci
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy.
| |
Collapse
|
35
|
Faria L, Canato S, Jesus TT, Gonçalves M, Guerreiro PS, Lopes CS, Meireles I, Morais-de-Sá E, Paredes J, Janody F. Activation of an actin signaling pathway in pre-malignant mammary epithelial cells by P-cadherin is essential for transformation. Dis Model Mech 2023; 16:dmm049652. [PMID: 36808468 PMCID: PMC9983776 DOI: 10.1242/dmm.049652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/19/2023] [Indexed: 02/23/2023] Open
Abstract
Alterations in the expression or function of cell adhesion molecules have been implicated in all steps of tumor progression. Among those, P-cadherin is highly enriched in basal-like breast carcinomas, playing a central role in cancer cell self-renewal, collective cell migration and invasion. To establish a clinically relevant platform for functional exploration of P-cadherin effectors in vivo, we generated a humanized P-cadherin Drosophila model. We report that actin nucleators, Mrtf and Srf, are main P-cadherin effectors in fly. We validated these findings in a human mammary epithelial cell line with conditional activation of the SRC oncogene. We show that, prior to promoting malignant phenotypes, SRC induces a transient increase in P-cadherin expression, which correlates with MRTF-A accumulation, its nuclear translocation and the upregulation of SRF target genes. Moreover, knocking down P-cadherin, or preventing F-actin polymerization, impairs SRF transcriptional activity. Furthermore, blocking MRTF-A nuclear translocation hampers proliferation, self-renewal and invasion. Thus, in addition to sustaining malignant phenotypes, P-cadherin can also play a major role in the early stages of breast carcinogenesis by promoting a transient boost of MRTF-A-SRF signaling through actin regulation.
Collapse
Affiliation(s)
- Lídia Faria
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Master Programme in Oncology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Sara Canato
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Physiology and Cancer Program, Champalimaud Foundation, Avenida de Brasília, 1400-038 Lisboa, Portugal
| | - Tito T. Jesus
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Margarida Gonçalves
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Patrícia S. Guerreiro
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Vector B2B - Drug Developing - Associação Para Investigação em Biotecnologia, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carla S. Lopes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Isabel Meireles
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Eurico Morais-de-Sá
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Joana Paredes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- FMUP, Medical Faculty of University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Florence Janody
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, P-2780-156 Oeiras, Portugal
| |
Collapse
|
36
|
Zhu T, Guo J, Wu Y, Lei T, Zhu J, Chen H, Kala S, Wong KF, Cheung CP, Huang X, Zhao X, Yang M, Sun L. The mechanosensitive ion channel Piezo1 modulates the migration and immune response of microglia. iScience 2023; 26:105993. [PMID: 36798430 PMCID: PMC9926228 DOI: 10.1016/j.isci.2023.105993] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/28/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Microglia are the brain's resident immune cells, performing surveillance to promote homeostasis and healthy functioning. While microglial chemical signaling is well-studied, mechanical cues regulating their function are less well-understood. Here, we investigate the role of the mechanosensitive ion channel Piezo1 in microglia migration, pro-inflammatory cytokine production, and stiffness sensing. In Piezo1 knockout transgenic mice, we demonstrated the functional expression of Piezo1 in microglia and identified genes whose expression was consequently affected. Functional assays revealed that Piezo1 deficiency in microglia enhanced migration toward amyloid β-protein, and decreased levels of pro-inflammatory cytokines produced upon stimulation by lipopolysaccharide, both in vitro and in vivo. The phenomenon could be mimicked or reversed chemically using a Piezo1-specific agonist or antagonist. Finally, we also showed that Piezo1 mediated the effect of substrate stiffness-induced migration and cytokine expression. Altogether, we show that Piezo1 is an important molecular mediator for microglia, its activation modulating microglial migration and immune responses.
Collapse
Affiliation(s)
- Ting Zhu
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Jinghui Guo
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Yong Wu
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Ting Lei
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Jiejun Zhu
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Hui Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China,Cell-gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shashwati Kala
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Kin Fung Wong
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Chi Pong Cheung
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Xiaohui Huang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Xinyi Zhao
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Minyi Yang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Lei Sun
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China,Corresponding author
| |
Collapse
|
37
|
EVL Promotes Osteo-/Odontogenic Differentiation of Dental Pulp Stem Cells via Activating JNK Signaling Pathway. Stem Cells Int 2023; 2023:7585111. [PMID: 36684389 PMCID: PMC9851786 DOI: 10.1155/2023/7585111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 12/06/2022] [Accepted: 12/20/2022] [Indexed: 01/14/2023] Open
Abstract
Objective Human dental pulp stem cells (hDPSCs) were recognized as a suitable and promising source of stem cells in dental pulp regeneration. However, the mechanism by which hDPSCs differentiation into osteo-/odontogenic lineage remains unclear. Ena/VASP-like protein (EVL) has been found to be involved in diverse biological processes. In this study, we explored the role and underlying mechanism of EVL in osteo-/odontogenic differentiation of hDPSCs. Methods Expression of EVL was detected in hDPSCs by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot (WB) analyses during osteo-/odontogenic differentiation. The function of EVL in osteo-/odontogenic differentiation and involvement of MAPK signaling pathways were evaluated by alkaline phosphatase (ALP) staining and activity, alizarin red staining (ARS), and qRT-PCR and western blot analyses. Results The expression of EVL was upregulated during osteo-/odontogenic differentiation of hDPSCs. Overexpression of EVL significantly increased osteo-/odontogenic capacity of hDPSCs, which was reflected in increased alkaline phosphatase (ALP) staining, ALP activity, mineralized nodule formation, and the expressions of genes related to osteo-/odontogenic differentiation, while downregulation of EVL inhibited it. In addition, EVL activated the JNK pathway and phosphorylation of p38 MAPK during differentiation procedure of hDPSCs. The EVL-enhanced differentiation of DPSCs was suppressed by blocking the JNK pathway, rather than the p38 MAPK pathway. Conclusion EVL promotes the osteo-/odontogenic differentiation of hDPSCs by activating the JNK pathway, providing a future target for osteo-/odontogenic differentiation and dental pulp regeneration.
Collapse
|
38
|
López-Cánovas JL, Hermán-Sánchez N, Del Rio-Moreno M, Fuentes-Fayos AC, Lara-López A, Sánchez-Frias ME, Amado V, Ciria R, Briceño J, de la Mata M, Castaño JP, Rodriguez-Perálvarez M, Luque RM, Gahete MD. PRPF8 increases the aggressiveness of hepatocellular carcinoma by regulating FAK/AKT pathway via fibronectin 1 splicing. Exp Mol Med 2023; 55:132-142. [PMID: 36609600 PMCID: PMC9898568 DOI: 10.1038/s12276-022-00917-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/18/2022] [Accepted: 11/08/2022] [Indexed: 01/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) pathogenesis is associated with alterations in splicing machinery components (spliceosome and splicing factors) and aberrant expression of oncogenic splice variants. We aimed to analyze the expression and potential role of the spliceosome component PRPF8 (pre-mRNA processing factor 8) in HCC. PRPF8 expression (mRNA/protein) was analyzed in a retrospective cohort of HCC patients (n = 172 HCC and nontumor tissues) and validated in two in silico cohorts (TCGA and CPTAC). PRPF8 expression was silenced in liver cancer cell lines and in xenograft tumors to understand the functional and mechanistic consequences. In silico RNAseq and CLIPseq data were also analyzed. Our results indicate that PRPF8 is overexpressed in HCC and associated with increased tumor aggressiveness (patient survival, etc.), expression of HCC-related splice variants, and modulation of critical genes implicated in cancer-related pathways. PRPF8 silencing ameliorated aggressiveness in vitro and decreased tumor growth in vivo. Analysis of in silico CLIPseq data in HepG2 cells demonstrated that PRPF8 binds preferentially to exons of protein-coding genes, and RNAseq analysis showed that PRPF8 silencing alters splicing events in multiple genes. Integrated and in vitro analyses revealed that PRPF8 silencing modulates fibronectin (FN1) splicing, promoting the exclusion of exon 40.2, which is paramount for binding to integrins. Consistent with this finding, PRPF8 silencing reduced FAK/AKT phosphorylation and blunted stress fiber formation. Indeed, HepG2 and Hep3B cells exhibited a lower invasive capacity in membranes treated with conditioned medium from PRPF8-silenced cells compared to medium from scramble-treated cells. This study demonstrates that PRPF8 is overexpressed and associated with aggressiveness in HCC and plays important roles in hepatocarcinogenesis by altering FN1 splicing, FAK/AKT activation and stress fiber formation.
Collapse
Affiliation(s)
- Juan L López-Cánovas
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain
| | - Natalia Hermán-Sánchez
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain
| | - Mercedes Del Rio-Moreno
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain
| | - Araceli Lara-López
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain
| | - Marina E Sánchez-Frias
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
| | - Víctor Amado
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Hepatic and Digestive Diseases (CIBERehd), 14004, Córdoba, Spain
| | - Rubén Ciria
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Unit of Hepatobiliary Surgery and Liver Transplantation, University Hospital Reina Sofia, 14004, Cordoba, Spain
| | - Javier Briceño
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Unit of Hepatobiliary Surgery and Liver Transplantation, University Hospital Reina Sofia, 14004, Cordoba, Spain
| | - Manuel de la Mata
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Hepatic and Digestive Diseases (CIBERehd), 14004, Córdoba, Spain
| | - Justo P Castaño
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain
| | - Manuel Rodriguez-Perálvarez
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Hepatic and Digestive Diseases (CIBERehd), 14004, Córdoba, Spain
| | - Raúl M Luque
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain
- Reina Sofía University Hospital, 14004, Córdoba, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain
| | - Manuel D Gahete
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), 14004, Córdoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004, Córdoba, Spain.
- Reina Sofía University Hospital, 14004, Córdoba, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), 14004, Córdoba, Spain.
| |
Collapse
|
39
|
Basu A, Paul MK, Weiss S. The actin cytoskeleton: Morphological changes in pre- and fully developed lung cancer. BIOPHYSICS REVIEWS 2022; 3:041304. [PMID: 38505516 PMCID: PMC10903407 DOI: 10.1063/5.0096188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 12/09/2022] [Indexed: 03/21/2024]
Abstract
Actin, a primary component of the cell cytoskeleton can have multiple isoforms, each of which can have specific properties uniquely suited for their purpose. These monomers are then bound together to form polymeric filaments utilizing adenosine triphosphate hydrolysis as a source of energy. Proteins, such as Arp2/3, VASP, formin, profilin, and cofilin, serve important roles in the polymerization process. These filaments can further be linked to form stress fibers by proteins called actin-binding proteins, such as α-actinin, myosin, fascin, filamin, zyxin, and epsin. These stress fibers are responsible for mechanotransduction, maintaining cell shape, cell motility, and intracellular cargo transport. Cancer metastasis, specifically epithelial mesenchymal transition (EMT), which is one of the key steps of the process, is accompanied by the formation of thick stress fibers through the Rho-associated protein kinase, MAPK/ERK, and Wnt pathways. Recently, with the advent of "field cancerization," pre-malignant cells have also been demonstrated to possess stress fibers and related cytoskeletal features. Analytical methods ranging from western blot and RNA-sequencing to cryo-EM and fluorescent imaging have been employed to understand the structure and dynamics of actin and related proteins including polymerization/depolymerization. More recent methods involve quantifying properties of the actin cytoskeleton from fluorescent images and utilizing them to study biological processes, such as EMT. These image analysis approaches exploit the fact that filaments have a unique structure (curvilinear) compared to the noise or other artifacts to separate them. Line segments are extracted from these filament images that have assigned lengths and orientations. Coupling such methods with statistical analysis has resulted in development of a new reporter for EMT in lung cancer cells as well as their drug responses.
Collapse
Affiliation(s)
| | | | - Shimon Weiss
- Author to whom correspondence should be addressed:
| |
Collapse
|
40
|
Merino-Casallo F, Gomez-Benito MJ, Hervas-Raluy S, Garcia-Aznar JM. Unravelling cell migration: defining movement from the cell surface. Cell Adh Migr 2022; 16:25-64. [PMID: 35499121 PMCID: PMC9067518 DOI: 10.1080/19336918.2022.2055520] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Cell motility is essential for life and development. Unfortunately, cell migration is also linked to several pathological processes, such as cancer metastasis. Cells' ability to migrate relies on many actors. Cells change their migratory strategy based on their phenotype and the properties of the surrounding microenvironment. Cell migration is, therefore, an extremely complex phenomenon. Researchers have investigated cell motility for more than a century. Recent discoveries have uncovered some of the mysteries associated with the mechanisms involved in cell migration, such as intracellular signaling and cell mechanics. These findings involve different players, including transmembrane receptors, adhesive complexes, cytoskeletal components , the nucleus, and the extracellular matrix. This review aims to give a global overview of our current understanding of cell migration.
Collapse
Affiliation(s)
- Francisco Merino-Casallo
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Maria Jose Gomez-Benito
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Silvia Hervas-Raluy
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
41
|
Han SJ, Kwon S, Kim KS. Contribution of mechanical homeostasis to epithelial-mesenchymal transition. Cell Oncol (Dordr) 2022; 45:1119-1136. [PMID: 36149601 DOI: 10.1007/s13402-022-00720-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Metastasis refers to the spread of cancer cells from a primary tumor to other parts of the body via the lymphatic system and bloodstream. With tremendous effort over the past decades, remarkable progress has been made in understanding the molecular and cellular basis of metastatic processes. Metastasis occurs through five steps, including infiltration and migration, intravasation, survival, extravasation, and colonization. Various molecular and cellular factors involved in the metastatic process have been identified, such as epigenetic factors of the extracellular matrix (ECM), cell-cell interactions, soluble signaling, adhesion molecules, and mechanical stimuli. However, the underlying cause of cancer metastasis has not been elucidated. CONCLUSION In this review, we have focused on changes in the mechanical properties of cancer cells and their surrounding environment to understand the causes of cancer metastasis. Cancer cells have unique mechanical properties that distinguish them from healthy cells. ECM stiffness is involved in cancer cell growth, particularly in promoting the epithelial-mesenchymal transition (EMT). During tumorigenesis, the mechanical properties of cancer cells change in the direction opposite to their environment, resulting in a mechanical stress imbalance between the intracellular and extracellular domains. Disruption of mechanical homeostasis may be one of the causes of EMT that triggers the metastasis of cancer cells.
Collapse
Affiliation(s)
- Se Jik Han
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul, Korea.,Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea
| | - Sangwoo Kwon
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
42
|
Pouliquen DL, Malloci M, Boissard A, Henry C, Guette C. Proteomes of Residual Tumors in Curcumin-Treated Rats Reveal Changes in Microenvironment/Malignant Cell Crosstalk in a Highly Invasive Model of Mesothelioma. Int J Mol Sci 2022; 23:ijms232213732. [PMID: 36430209 PMCID: PMC9691155 DOI: 10.3390/ijms232213732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Curcumin exhibits both immunomodulatory properties and anticarcinogenic effects which have been investigated in different experimental tumor models and cancer types. Its interactions with multiple signaling pathways have been documented through proteomic studies on malignant cells in culture; however, in vivo approaches are scarce. In this study, we used a rat model of highly invasive peritoneal mesothelioma to analyze the residual tumor proteomes of curcumin-treated rats in comparison with untreated tumor-bearing rats (G1) and provide insights into the modifications in the tumor microenvironment/malignant cell crosstalk. The cross-comparing analyses of the histological sections of residual tumors from two groups of rats given curcumin twice on days 21 and 26 after the tumor challenge (G2) or four times on days 7, 9, 11 and 14 (G3), in comparison with G1, identified a common increase in caveolin-1 which linked with significant abundance changes affecting 115 other proteins. The comparison of G3 vs. G2 revealed additional features for 65 main proteins, including an increase in histidine-rich glycoprotein and highly significant abundance changes for 22 other proteins regulating the tumor microenvironment, linked with the presence of numerous activated T cells. These results highlight new features in the multiple actions of curcumin on tumor microenvironment components and cancer cell invasiveness.
Collapse
Affiliation(s)
- Daniel L. Pouliquen
- Université d’Angers, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
- Correspondence: ; Tel.: +33-2-41352854
| | - Marine Malloci
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, F-44000 Nantes, France
| | - Alice Boissard
- Université d’Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Cécile Henry
- Université d’Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Catherine Guette
- Université d’Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| |
Collapse
|
43
|
Weber A, Benitez R, Toca‐Herrera JL. Measuring biological materials mechanics with atomic force microscopy - Determination of viscoelastic cell properties from stress relaxation experiments. Microsc Res Tech 2022; 85:3284-3295. [PMID: 35736395 PMCID: PMC9796732 DOI: 10.1002/jemt.24184] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 01/07/2023]
Abstract
Cells are complex, viscoelastic bodies. Their mechanical properties are defined by the arrangement of semiflexible cytoskeletal fibers, their crosslinking, and the active remodeling of the cytoskeletal network. Atomic force microscopy (AFM) is an often-used technique for the study of cell mechanics, enabling time- and frequency-dependent measurements with nanometer resolution. Cells exhibit time-dependent deformation when stress is applied. In this work, we have investigated the stress relaxation of HeLa cells when subjected to a constant strain. We have varied the applied force (1, 2, 4, and 8 nN) and pause time (1, 10, and 60 s) to check for common assumptions for the use of models of linear viscoelasticity. Then, we have applied three models (standard linear solid, five element Maxwell, power law rheology) to study their suitability to fit the datasets. We show that the five element Maxwell model captures the stress relaxation response the best while still retaining a low number of free variables. This work serves as an introduction and guide when performing stress relaxation experiments on soft matter using AFM. RESEARCH HIGHLIGHTS: Cells exhibit linear viscoelastic properties when subjected to stress relaxation measurements at the studied different forces and times. The stress relaxation is best described by a five element Maxwell model. All three used models capture a softening and fluidization of cells when disrupting actin filaments.
Collapse
Affiliation(s)
- Andreas Weber
- Institute of Biophysics, Department of NanobiotechnologyUniversity of Natural Resources and Life Sciences Vienna (BOKU)ViennaAustria
| | - Rafael Benitez
- Departamento de Matemáticas para la Economía y la EmpresaFacultad de Economía, Universidad de ValenciaValenciaSpain
| | - José L. Toca‐Herrera
- Institute of Biophysics, Department of NanobiotechnologyUniversity of Natural Resources and Life Sciences Vienna (BOKU)ViennaAustria
| |
Collapse
|
44
|
Liu G, Li J, Wu C. Reciprocal regulation of actin filaments and cellular metabolism. Eur J Cell Biol 2022; 101:151281. [PMID: 36343493 DOI: 10.1016/j.ejcb.2022.151281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 09/23/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
For cells to adhere, migrate and proliferate, remodeling of the actin cytoskeleton is required. This process consumes a large amount of ATP while having an intimate connection with cellular metabolism. Signaling pathways that regulate energy homeostasis can also affect actin dynamics, whereas a variety of actin binding proteins directly or indirectly interact with the anabolic and catabolic regulators in cells. Here, we discuss the inter-regulation between actin filaments and cellular metabolism, reviewing recent discoveries on key metabolic enzymes that respond to actin remodeling as well as historical findings on metabolic stress-induced cytoskeletal reorganization. We also address emerging techniques that would benefit the study of cytoskeletal dynamics and cellular metabolism in high spatial-temporal resolution.
Collapse
Affiliation(s)
- Geyao Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jiayi Li
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; International Cancer Institute, Peking University, Beijing 100191, China.
| |
Collapse
|
45
|
Tang S, Liu W, Yong L, Liu D, Lin X, Huang Y, Wang H, Cai F. Reduced Expression of KRT17 Predicts Poor Prognosis in HER2high Breast Cancer. Biomolecules 2022; 12:biom12091183. [PMID: 36139022 PMCID: PMC9496156 DOI: 10.3390/biom12091183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer (BC) is one of the most common types of malignancies in women and greatly threatens female health. KRT17 is a member of the keratin (KRT) protein family that is abundant in the outer layer of the skin, where it protects epithelial cells from damage. Although KRT17 has been studied in many types of cancer, the expression of KRT17 in specific subtypes of BC remains to be determined. In our study, we explored the expression and prognostic implications of KRT17 in BC patients using mRNA transcriptome data and clinical BC data from The Cancer Genome Atlas (TCGA). Receiver operating characteristic (ROC) curves and the chi-square test were used to assess the diagnostic value of KRT17 expression. Quantitative real-time PCR (qRT−PCR) analysis of BC cells and tissues and immunohistochemistry (IHC) analysis of clinical tissues were used for external validation. Furthermore, the relationship between KRT17 and immune function was studied by using the CIBERSORT algorithm to predict the proportions of tumor-infiltrating immune cells (TIICs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to explore the potential mechanisms by which KRT17 expression influences patient survival. We found that KRT17 expression was significantly lower in BC tissues than in normal tissues, especially in the luminal-A, luminal-B and human epidermal growth factor receptor-2 (HER2)+ subtypes of BC. ROC analysis revealed that KRT17 expression had moderate diagnostic value. Interestingly, decreased expression of KRT17 was significantly correlated with poor prognosis in BC patients, especially in HER2high and ERhigh patients. This trend was also verified by tissue microarray (TMA) analysis. KRT17 was found to be involved in some antitumor immune pathways, especially the IL-17 signaling pathway, and associated with multiple immune cells, such as natural killer (NK) and CD4+ T cells. In conclusion, high expression of KRT17 predicted favorable prognosis in BC patients with higher HER2 expression. This result may indicate that KRT17 plays a different role depending on the level of HER2 expression and could serve as a promising and sensitive biomarker for the diagnosis and prognostication of HER2high BC.
Collapse
Affiliation(s)
- Shasha Tang
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, No.450 Tengyue Road, Shanghai 200090, China
| | - Wenjing Liu
- Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 201100, China
| | - Liyun Yong
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, No.450 Tengyue Road, Shanghai 200090, China
| | - Dongyang Liu
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, No.450 Tengyue Road, Shanghai 200090, China
| | - Xiaoyan Lin
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, No.450 Tengyue Road, Shanghai 200090, China
| | - Yuan Huang
- Cellomics International Limited, Hong Kong, China
| | - Hui Wang
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, No.279 Zhouzhu Highway, Shanghai 201318, China
- Correspondence: (H.W.); (F.C.)
| | - Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, No.450 Tengyue Road, Shanghai 200090, China
- Correspondence: (H.W.); (F.C.)
| |
Collapse
|
46
|
Biomechanics of cancer stem cells. Essays Biochem 2022; 66:359-369. [PMID: 35942932 DOI: 10.1042/ebc20220014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs) have been believed to be one driving force for tumor progression and drug resistance. Despite the significance of biochemical signaling in malignancy, highly malignant tumor cells or CSCs exhibit lower cellular stiffness than weakly malignant cells or non-CSCs, which are softer than their healthy counterparts, suggesting the inverse correlation between cell stiffness and malignancy. Recent years have witnessed the rapid accumulation of evidence illustrating the reciprocity between cell cytoskeleton/mechanics and CSC functions and the potential of cellular stiffness for specific targeting of CSCs. However, a systematic understanding of tumor cell mechanics and their role in CSCs and tumor progression is still lacking. The present review summarizes the recent progress in the alterations of tumor cell cytoskeleton and stiffness at different stages of tumor progression and recapitulates the relationship between cellular stiffness and CSC functions. The altered cell mechanics may mediate the mechanoadaptive responses that possibly empower CSCs to survive and thrive during metastasis. Furthermore, we highlight the possible impact of tumor cell mechanics on CSC malignancy, which may potentiate low cell stiffness as a mechanical marker for CSC targeting.
Collapse
|
47
|
Khalil K, Eon A, Janody F. Cell Architecture-Dependent Constraints: Critical Safeguards to Carcinogenesis. Int J Mol Sci 2022; 23:8622. [PMID: 35955754 PMCID: PMC9369145 DOI: 10.3390/ijms23158622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Animal cells display great diversity in their shape. These morphological characteristics result from crosstalk between the plasma membrane and the force-generating capacities of the cytoskeleton macromolecules. Changes in cell shape are not merely byproducts of cell fate determinants, they also actively drive cell fate decisions, including proliferation and differentiation. Global and local changes in cell shape alter the transcriptional program by a multitude of mechanisms, including the regulation of physical links between the plasma membrane and the nuclear envelope and the mechanical modulation of cation channels and signalling molecules. It is therefore not surprising that anomalies in cell shape contribute to several diseases, including cancer. In this review, we discuss the possibility that the constraints imposed by cell shape determine the behaviour of normal and pro-tumour cells by organizing the whole interconnected regulatory network. In turn, cell behaviour might stabilize cells into discrete shapes. However, to progress towards a fully transformed phenotype and to acquire plasticity properties, pro-tumour cells might need to escape these cell shape restrictions. Thus, robust controls of the cell shape machinery may represent a critical safeguard against carcinogenesis.
Collapse
Affiliation(s)
- Komal Khalil
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Master Programme in Oncology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Alice Eon
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Magistère Européen de Génétique, Université Paris Cité, 5 Rue Thomas Mann, 75013 Paris, France
| | - Florence Janody
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| |
Collapse
|
48
|
Senigagliesi B, Samperi G, Cefarin N, Gneo L, Petrosino S, Apollonio M, Caponnetto F, Sgarra R, Collavin L, Cesselli D, Casalis L, Parisse P. Triple negative breast cancer-derived small extracellular vesicles as modulator of biomechanics in target cells. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 44:102582. [PMID: 35817390 DOI: 10.1016/j.nano.2022.102582] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 12/14/2022]
Abstract
Extracellular vesicle (EV) mediated communication has recently been proposed as one of the pivotal routes in the development of cancer metastasis. EVs are nano-sized vesicles swapped between cells, carrying a biologically active content that can promote tumor-induced immune suppression, metastasis and angiogenesis. Thus, EVs constitute a potential target in cancer therapy. However, their role in triggering the premetastatic niche and in tumor spreading is still unclear. Here, we focused on the EV ability to modulate the biomechanical properties of target cells, known to play a crucial role in metastatic spreading. To this purpose, we isolated and thoroughly characterized triple-negative breast cancer (TNBC)-derived small EVs. We then evaluated variations in the mechanical properties (cell stiffness, cytoskeleton/nuclear/morphology and Yap activity rearrangements) of non-metastatic breast cancer MCF7 cells upon EV treatment. Our results suggest that TNBC-derived small EVs are able to directly modify MCF7 cells by inducing a decrease in cell stiffness, rearrangements in cytoskeleton, focal adhesions and nuclear/cellular morphology, and an increase in Yap downstream gene expression. Testing the biomechanical response of cells after EV addition might represent a new functional assay in metastatic cancer framework that can be exploited for future application both in diagnosis and in therapy.
Collapse
Affiliation(s)
- Beatrice Senigagliesi
- Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy; Elettra-Sincrotrone Trieste S.C.p.A, Trieste, Italy.
| | | | - Nicola Cefarin
- Istituto Officina dei Materiali Consiglio Nazionale delle Ricerche, Trieste, Italy
| | | | - Sara Petrosino
- Telethon Institute of Genetics and Medicine, Naples, Italy.
| | - Mattia Apollonio
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Trieste, Italy.
| | - Licio Collavin
- Department of Life Sciences, University of Trieste, Trieste, Italy.
| | - Daniela Cesselli
- Pathology Department, University Hospital of Udine, Udine, Italy; Department of Medicine, University of Udine, Udine, Italy.
| | | | - Pietro Parisse
- Elettra-Sincrotrone Trieste S.C.p.A, Trieste, Italy; Istituto Officina dei Materiali Consiglio Nazionale delle Ricerche, Trieste, Italy.
| |
Collapse
|
49
|
Gao S, Wang S, Zhao Z, Zhang C, Liu Z, Ye P, Xu Z, Yi B, Jiao K, Naik GA, Wei S, Rais-Bahrami S, Bae S, Yang WH, Sonpavde G, Liu R, Wang L. TUBB4A interacts with MYH9 to protect the nucleus during cell migration and promotes prostate cancer via GSK3β/β-catenin signalling. Nat Commun 2022; 13:2792. [PMID: 35589707 PMCID: PMC9120517 DOI: 10.1038/s41467-022-30409-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/28/2022] [Indexed: 01/22/2023] Open
Abstract
Human tubulin beta class IVa (TUBB4A) is a member of the β-tubulin family. In most normal tissues, expression of TUBB4A is little to none, but it is highly expressed in human prostate cancer. Here we show that high expression levels of TUBB4A are associated with aggressive prostate cancers and poor patient survival, especially for African-American men. Additionally, in prostate cancer cells, TUBB4A knockout (KO) reduces cell growth and migration but induces DNA damage through increased γH2AX and 53BP1. Furthermore, during constricted cell migration, TUBB4A interacts with MYH9 to protect the nucleus, but either TUBB4A KO or MYH9 knockdown leads to severe DNA damage and reduces the NF-κB signaling response. Also, TUBB4A KO retards tumor growth and metastasis. Functional analysis reveals that TUBB4A/GSK3β binds to the N-terminal of MYH9, and that TUBB4A KO reduces MYH9-mediated GSK3β ubiquitination and degradation, leading to decreased activation of β-catenin signaling and its relevant epithelial-mesenchymal transition. Likewise, prostate-specific deletion of Tubb4a reduces spontaneous tumor growth and metastasis via inhibition of NF-κB, cyclin D1, and c-MYC signaling activation. Our results suggest an oncogenic role of TUBB4A and provide a potentially actionable therapeutic target for prostate cancers with TUBB4A overexpression. The β-tubulin family protein TUBB4A is highly expressed in cancer but it’s molecular role is unclear. Here, the authors show that TUBB4A is required to protect the nucleus from genomic instability during migration and that it’s over expression promotes cancer progression.
Collapse
Affiliation(s)
- Song Gao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shuaibin Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhiying Zhao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chao Zhang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhicao Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ping Ye
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhifang Xu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Baozhu Yi
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gurudatta A Naik
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shi Wei
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Soroush Rais-Bahrami
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sejong Bae
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei-Hsiung Yang
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | | | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
50
|
Basu A, Paul MK, Alioscha-Perez M, Grosberg A, Sahli H, Dubinett SM, Weiss S. Statistical parametrization of cell cytoskeleton reveals lung cancer cytoskeletal phenotype with partial EMT signature. Commun Biol 2022; 5:407. [PMID: 35501466 PMCID: PMC9061773 DOI: 10.1038/s42003-022-03358-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 04/12/2022] [Indexed: 12/14/2022] Open
Abstract
Epithelial–mesenchymal Transition (EMT) is a multi-step process that involves cytoskeletal rearrangement. Here, developing and using an image quantification tool, Statistical Parametrization of Cell Cytoskeleton (SPOCC), we have identified an intermediate EMT state with a specific cytoskeletal signature. We have been able to partition EMT into two steps: (1) initial formation of transverse arcs and dorsal stress fibers and (2) their subsequent conversion to ventral stress fibers with a concurrent alignment of fibers. Using the Orientational Order Parameter (OOP) as a figure of merit, we have been able to track EMT progression in live cells as well as characterize and quantify their cytoskeletal response to drugs. SPOCC has improved throughput and is non-destructive, making it a viable candidate for studying a broad range of biological processes. Further, owing to the increased stiffness (and by inference invasiveness) of the intermediate EMT phenotype compared to mesenchymal cells, our work can be instrumental in aiding the search for future treatment strategies that combat metastasis by specifically targeting the fiber alignment process. A computational method for automated quantification of actin stress fiber alignment in fluorescence images of cultured cells is presented, used to detect changes in stress fiber organization during EMT, with pathways regulating actin dynamics manipulated leading to the discovery of a cytoskeletal phenotype.
Collapse
Affiliation(s)
- Arkaprabha Basu
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Manash K Paul
- Department of Medicine, University of California Los Angeles, Los Angles, CA, USA.,Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Mitchel Alioscha-Perez
- Electronics and Informatics Department, Vrije Universiteit Brussel, Brussels, Belgium.,Interuniversity Microelectronics Centre, Heverlee, Belgium
| | - Anna Grosberg
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.,The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, CA, USA
| | - Hichem Sahli
- Electronics and Informatics Department, Vrije Universiteit Brussel, Brussels, Belgium.,Interuniversity Microelectronics Centre, Heverlee, Belgium
| | - Steven M Dubinett
- Department of Medicine, University of California Los Angeles, Los Angles, CA, USA.,Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,California NanoSystems Institute, Los Angeles, CA, USA.,VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Shimon Weiss
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA. .,California NanoSystems Institute, Los Angeles, CA, USA. .,Department of Physiology, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|