1
|
Zhang X, He X, Si Y, Nie X, Lun S, Wang C, Lin L, Yan Y. A dual sensitive fluorescence probe for the simultaneous visualization of hypochlorite and viscosity in living cells and zebrafish. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 325:125149. [PMID: 39293203 DOI: 10.1016/j.saa.2024.125149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
As vital microenvironmental indicators, hypochlorite and viscosity levels can be applied to diagnose diseases. Thus, real time and in situ monitoring of hypochlorite and viscosity has gained increasing attentions. A fluorescence probe named JDK was developed by using a novel acceptor of 2-(2-(5-(dimethylamino)thiophen-2-yl)vinyl)-1,3,3-trimethyl-3H-indol-1-ium fluorophore basing on FRET and TICT system. JDK possessed large Stokes shift, broad emission peak gap, prominent photostability and biocompatibility. JDK could track hypochlorite with rapid response time (within 1 min) and low detection limit (28.5 nM). Meaningfully, JDK was favor to append on mitochondria where physiological processes can be disrupted deriving from the abnormal concentrations of hypochlorite and viscosity. Successfully, JDK could visualize endogenous hypochlorite and viscosity in living cells and zebrafish.
Collapse
Affiliation(s)
- Xin Zhang
- School of Public Health, Jining Medical University, Jining, Shandong, 272067, PR China; School of Public Health, Shandong Second Medical University, Weifang 261053, PR China
| | - Xiaoying He
- Department of Nephrology, Shanxi Provincial People's Hospital, Taiyuan, PR China
| | - Yanmei Si
- School of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, 272067, PR China
| | - Xuqing Nie
- School of Public Health, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Shenghui Lun
- School of Public Health, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Changqin Wang
- School of Public Health, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Li Lin
- School of Public Health, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Yehao Yan
- School of Public Health, Jining Medical University, Jining, Shandong, 272067, PR China.
| |
Collapse
|
2
|
Yao L, Li Y. Implementation of actin polymerization and depolymerization in a two-dimensional cell migration model and its implications on mammalian cell morphology and velocity. J Theor Biol 2025; 596:111977. [PMID: 39510349 DOI: 10.1016/j.jtbi.2024.111977] [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: 03/13/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024]
Abstract
Cell migration, a pivotal process in wound healing, immune response, and even cancer metastasis, manifests through intricate interplay between morphology, speed, and cytoskeletal dynamics. Mathematical modeling emerges as a powerful tool to dissect these complex interactions. This work presents a two-dimensional immersed boundary model for mammalian cell migration, incorporating both filamentous actin (F-actin) and monomeric actin (G-actin) to explicitly capture polymerization and depolymerization. This model builds upon our previous one-dimensional efforts, now enabling us to explore the impact of G-actin on not just cell velocity but also morphology. We compare predictions from both models, revealing that while the one-dimensional model captures core dynamics along the cell's axis, the two-dimensional model excels in portraying cell shape evolution and transverse variations in actin concentration and velocity. Our findings highlight the crucial role of including G-actin in shaping cell morphology. Actin velocity aligned with migration direction elongates the cell, while velocity normal to the membrane promotes spreading. Importantly, the model establishes a link between these microscopic aspects and macroscopic observables like cell shape, offering a deeper understanding of cell migration dynamics. This work not only provides a more comprehensive picture of cell migration but also paves the way for future studies exploring the interplay of actin dynamics, cell morphology, and biophysical parameters in diverse biological contexts.
Collapse
Affiliation(s)
- Lingxing Yao
- Department of Mathematics, University of Akron, Akron, OH 44325, USA
| | - Yizeng Li
- Department of Biomedical Engineering, Binghamton University, SUNY, Binghamton, NY 13902, USA.
| |
Collapse
|
3
|
Amitrano A, Yuan Q, Agarwal B, Sen A, Dance YW, Zuo Y, Phillip JM, Gu L, Konstantopoulos K. Extracellular fluid viscosity regulates human mesenchymal stem cell lineage and function. SCIENCE ADVANCES 2025; 11:eadr5023. [PMID: 39742493 DOI: 10.1126/sciadv.adr5023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
Abstract
Human mesenchymal stem cells (hMSCs) respond to mechanical stimuli, including stiffness and viscoelasticity. To date, it is unknown how extracellular fluid viscosity affects hMSC function on substrates of different stiffness and viscoelasticity. While hMSCs assume an adipogenic phenotype on gels of low stiffness and prescribed stress relaxation times, elevated fluid viscosity is sufficient to bias hMSCs toward an osteogenic phenotype. Elevated viscosity induces Arp2/3-dependent actin remodeling, enhances NHE1 activity, and promotes hMSC spreading via up-regulation of integrin-linked kinase. The resulting increase in membrane tension triggers the activation of transient receptor potential cation vanilloid 4 to facilitate calcium influx, thereby stimulating RhoA/ROCK and driving YAP-dependent RUNX2 translocation to the nucleus, leading to osteogenic differentiation. hMSCs on soft gels at elevated relative to basal viscosity favor an M2 macrophage phenotype. This study establishes fluid viscosity as a key physical cue that imprints osteogenic memory in hMSCs and promotes an immunosuppressive phenotype.
Collapse
Affiliation(s)
- Alice Amitrano
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Qinling Yuan
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bhawana Agarwal
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anindya Sen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yoseph W Dance
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yi Zuo
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Deparment of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jude M Phillip
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | - Luo Gu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Deparment of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| |
Collapse
|
4
|
Bayer Kömüşdoğan E, Batool S, Şahin E, Yildirim E, Işık M, Tanyeli C. Multicomponent synthesis of stereogenic-at-boron fluorophores (BOSPYR) from boronic acids, salicylaldehydes, and 2-formylpyrrole hydrazones. Chem Commun (Camb) 2025; 61:576-579. [PMID: 39656116 DOI: 10.1039/d4cc03956d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
This work describes one-step syntheses of various stereogenic-at-boron fluorochromes (BOSPYR) via multicomponent reactions involving readily accessible boronic acids, salicylaldehydes, and 2-formylpyrrole hydrazones. The dyes absorb and emit in the visible region of the electromagnetic radiation, and are characterized by large Stokes shifts (2850-4930 cm-1) with weak fluorescence emissions (Φfl: 1.5-9.1%). Notably, the dimmed fluorescence of BOSPYRs recovers upon transition to viscous media (21-fold for 1a). The representative compound 1a exhibits clear Cotton effects with dissymmetry factors of ca. |gabs| ∼ 1.9 × 10-3 in the visible region, indicating efficient asymmetry induction to the chromophore. The X-ray molecular structure of 1a shows that the chromophore deviates from planarity by 17.2°, which may contribute significantly to the inherent chirality of the fluorophore. A computational examination of excited states by time-dependent density functional theory (TD-DFT) identifies the emission mechanism as arising from a locally-excited (LE) state.
Collapse
Affiliation(s)
| | - Sania Batool
- Department of Chemistry, Middle East Technical University, 06800 Ankara, Turkey.
| | - Ertan Şahin
- Department of Chemistry, Atatürk University, Erzurum, 25240, Turkey
| | - Erol Yildirim
- Department of Chemistry, Middle East Technical University, 06800 Ankara, Turkey.
| | - Murat Işık
- Department of Food Engineering, Bingöl University, Bingöl, 12000, Turkey.
| | - Cihangir Tanyeli
- Department of Chemistry, Middle East Technical University, 06800 Ankara, Turkey.
| |
Collapse
|
5
|
Junqueira Alves C, Hannah T, Sadia S, Kolsteeg C, Dixon A, Wiener RJ, Nguyen H, Tipping MJ, Silva Ladeira J, Fernandes da Costa Franklin P, de Paula Dutra de Nigro N, Alves Dias R, Zabala Capriles PV, Rodrigues Furtado de Mendonça JP, Slesinger PA, Costa KD, Zou H, Friedel RH. Invasion of glioma cells through confined space requires membrane tension regulation and mechano-electrical coupling via Plexin-B2. Nat Commun 2025; 16:272. [PMID: 39747004 PMCID: PMC11697315 DOI: 10.1038/s41467-024-55056-6] [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: 02/28/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Glioblastoma (GBM) is a malignant brain tumor with diffuse infiltration. Here, we demonstrate how GBM cells usurp guidance receptor Plexin-B2 for confined migration through restricted space. Using live-cell imaging to track GBM cells negotiating microchannels, we reveal endocytic vesicle accumulation at cell front and filamentous actin assembly at cell rear in a polarized manner. These processes are interconnected and require Plexin-B2 signaling. We further show that Plexin-B2 governs membrane tension and other membrane features such as endocytosis, phospholipid composition, and inner leaflet surface charge, thus providing biophysical mechanisms by which Plexin-B2 promotes GBM invasion. Together, our studies unveil how GBM cells regulate membrane tension and mechano-electrical coupling to adapt to physical constraints and achieve polarized confined migration.
Collapse
Affiliation(s)
- Chrystian Junqueira Alves
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - Theodore Hannah
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sita Sadia
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christy Kolsteeg
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Angela Dixon
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Robert J Wiener
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ha Nguyen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Murray J Tipping
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Júlia Silva Ladeira
- Department of Computer Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | | | - Rodrigo Alves Dias
- Department of Physics, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | | | - Paul A Slesinger
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kevin D Costa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - Roland H Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
6
|
Rosalina R, Weerapreeyakul N, Sutthanut K, Kamwilaisak K, Sakonsinsiri C. Nanocellulose-based Pickering emulsion of sesamolin manifested increased anticancer activity and necrosis in human colon cancer (HCT116) cells. Int J Biol Macromol 2024; 292:139225. [PMID: 39732237 DOI: 10.1016/j.ijbiomac.2024.139225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Sesamolin possesses limited aqueous solubility, a drawback for biological activity study in cancer cell models. This study aimed to enhance sesamolin's ability to fight cancer, as it is a bioactive compound with low water solubility found in sesame. We developed different Pickering emulsion delivery systems and tested their anticancer effects on various cancer cell types. Sesamolin was incorporated into either sesame or olive oil and subsequently formulated as oil in water (o/w) Pickering emulsions stabilized by the carboxylated cellulose nanocrystal (cCNC). The anticancer activity was determined based on cell viability and the induction of cell death mechanisms. The results demonstrated a synergistic effect of the components in the emulsion, including sesamolin, sesame oil, and olive oil, and a decrease in HCT116 viability in a concentration-dependent manner and selectively on cancer cells compared to non-cancerous Vero cells. The primary mode of cell death was predominantly ROS-induced necrosis, with no change in caspase 3/7 activity, indicating the absence of apoptosis. This study first presents the necrotic cell death mechanism induced by sesamolin. The findings reveal that the cCNC emulsion delivery system is safe and appropriate for transporting lipophilic chemicals and can overcome solubility limitations.
Collapse
Affiliation(s)
- Reny Rosalina
- Graduate School (Biomedical Sciences Program), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Khaetthareeya Sutthanut
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Khanita Kamwilaisak
- Department of Chemical Engineering, Faculty of Engineering, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chadamas Sakonsinsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
7
|
Ni Q, Ge Z, Li Y, Shatkin G, Fu J, Sen A, Bera K, Yang Y, Wang Y, Wu Y, Nogueira Vasconcelos AC, Yan Y, Lin D, Feinberg AP, Konstantopoulos K, Sun SX. Cytoskeletal activation of NHE1 regulates mechanosensitive cell volume adaptation and proliferation. Cell Rep 2024; 43:114992. [PMID: 39579355 DOI: 10.1016/j.celrep.2024.114992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/24/2024] [Accepted: 11/04/2024] [Indexed: 11/25/2024] Open
Abstract
Mammalian cells rapidly respond to environmental changes by altering transmembrane water and ion fluxes, changing cell volume. Contractile forces generated by actomyosin have been proposed to mechanically regulate cell volume. However, our findings reveal a different mechanism in adherent cells, where elevated actomyosin activity increases cell volume in normal-like cells (NIH 3T3 and others) through interaction with the sodium-hydrogen exchanger isoform 1 (NHE1). This leads to a slow secondary volume increase (SVI) following the initial regulatory volume decrease during hypotonic shock. The active cell response is further confirmed by intracellular alkalinization during mechanical stretch. Moreover, cytoskeletal activation of NHE1 during SVI deforms the nucleus, causing immediate transcriptomic changes and ERK-dependent growth inhibition. Notably, SVI and its associated changes are absent in many cancer cell lines or cells on compliant substrates with reduced actomyosin activity. Thus, actomyosin acts as a sensory element rather than a force generator during adaptation to environmental challenges.
Collapse
Affiliation(s)
- Qin Ni
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhuoxu Ge
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yizeng Li
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
| | - Gabriel Shatkin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jinyu Fu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Physics, Johns Hopkins University, Baltimore, MD, USA
| | - Anindya Sen
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Kaustav Bera
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yuhan Yang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yichen Wang
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yufei Wu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ana Carina Nogueira Vasconcelos
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yuqing Yan
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Dingchang Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew P Feinberg
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Konstantinos Konstantopoulos
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sean X Sun
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
8
|
Mallon CJ, Hassani M, Osofsky EH, Familo SB, Fenlon EE, Tucker MJ. Unraveling Hydration Shell Dynamics and Viscosity Effects Around Cyanamide Probes via 2D IR Spectroscopy. J Am Chem Soc 2024. [PMID: 39701978 DOI: 10.1021/jacs.4c12716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Hydration dynamics and solvent viscosity play critical roles in the structure and function of biomolecules. An overwhelming body of evidence suggests that protein and membrane fluctuations are closely linked to solvent fluctuations. While extensive research exists on the use of vibrational probes to detect local interactions and solvent dynamics, fewer studies have explored how the behavior of these reporters changes in response to bulk viscosity. To address this gap, two-dimensional infrared spectroscopy (2D IR) was employed in this study to investigate the ultrafast hydration dynamics around a cyanamide (NCN) probe attached to a nucleoside, deoxycytidine, in aqueous solutions with varying glycerol content. The use of a small vibrational probe on a targeted nucleic acid offers the potential to capture more localized hydration dynamics than alternative methods. The time scales for the frequency correlation decays were found to increase linearly with bulk viscosity, ranging from 0.9 to 11.4 ps over viscosities of 0.96-49.1 cP. Additionally, molecular dynamics (MD) simulations were performed to model the local hydration dynamics around the NCN probe. Interestingly, increasing the glycerol content did not significantly alter the hydration of the deoxycytidine. The MD simulations further suggested that the NCN probe's frequency fluctuations were primarily influenced by the dynamics of water in the second solvation shell. Cage correlation functions, which measure the movement of water molecules in and out of the second solvation shell, exhibited decays that closely matched those of the frequency-fluctuation correlation function (FFCF). These findings offer new insights into hydration dynamics and the impact of viscosity on biological systems.
Collapse
Affiliation(s)
- Christopher J Mallon
- Department of Chemistry, University of Nevada, Reno, 1664 N. Virginia Street, Reno, Nevada 89557, United States
| | - Majid Hassani
- Department of Chemistry, University of Nevada, Reno, 1664 N. Virginia Street, Reno, Nevada 89557, United States
| | - Ellia H Osofsky
- Department of Chemistry, Franklin & Marshall College, Lancaster, Pennsylvania 17604-3003, United States
| | - Savannah B Familo
- Department of Chemistry, Franklin & Marshall College, Lancaster, Pennsylvania 17604-3003, United States
| | - Edward E Fenlon
- Department of Chemistry, Franklin & Marshall College, Lancaster, Pennsylvania 17604-3003, United States
| | - Matthew J Tucker
- Department of Chemistry, University of Nevada, Reno, 1664 N. Virginia Street, Reno, Nevada 89557, United States
| |
Collapse
|
9
|
Mei J, Jiang J, Li Z, Pan Y, Xu K, Gao X, Yuan J, Li L, Wang Y, Wang L, Zhao A, Jiang S, Wang X, Yi S, Li S, Xue Y, Ma Y, Liu Y, Wang Y, Li J, Chen C, Liu Y. Increased perfluorooctanoic acid accumulation facilitates the migration and invasion of lung cancer cells via remodeling cell mechanics. Proc Natl Acad Sci U S A 2024; 121:e2408575121. [PMID: 39665760 DOI: 10.1073/pnas.2408575121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Perfluoroalkyl and polyfluoroalkyl substances (PFAS) are widely used in industrial and household products, raising serious concerns due to their environmental persistence and mobility. Epidemiological studies have reported potential carcinogenic risks of PFAS based on their widespread occurrence and population exposure. In this study, we observed that perfluorooctanoic acid (PFOA), a common PFAS, functions as a mechanical regulator in lung cancer cells. PFOA exposure reduces cell stiffness, thereby decreasing cell adhesion and enhancing immune evasion, ultimately exacerbating tumor metastasis. In various lung cancer models, more aggressive tumor metastases have been observed in the PFOA exposure group. Additionally, serum PFOA levels in patients with advanced lung adenocarcinoma were significantly higher than those in patients with early-stage disease. Mechanistically, the interaction between PFOA and transmembrane integrins in cancer cells triggers changes in cellular mechanical properties, leading to the reorganization of the cytoskeleton, and activation of the intracellular FAK-PI3K-Akt signaling pathway. Our findings demonstrate that in individuals with lung adenocarcinoma, PFOA can increase the risk of cancer metastasis even at daily exposure levels.
Collapse
Affiliation(s)
- Jie Mei
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
- School of Nano Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jipeng Jiang
- Department of Thoracic Surgery, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Zhao Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yue Pan
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
| | - Ke Xu
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
- School of Nano Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinglong Gao
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
- School of Nano Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Yuan
- Department of Pathology, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Lili Li
- National Institutes for Food and Drug Control, Beijing 102629, China
| | - Yufei Wang
- Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100039, China
| | - Liuxiang Wang
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
| | - Ailin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Shasha Jiang
- Department of Thoracic Surgery, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Xinlian Wang
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
- School of Nano Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shaoqiong Yi
- National Center for Protein Sciences, Beijing 102206, China
| | - Shilin Li
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
| | - Yueguang Xue
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
| | - Yongfu Ma
- Department of Thoracic Surgery, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Yawei Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Juan Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Chunying Chen
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
- School of Nano Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Liu
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100190, China
- School of Nano Science and Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Christgen M, Caetano RA, Eisenburger M, Traulsen A, Altrock PM. Deficient cell-cell cohesion is linked with lobular localization in simplified models of lobular carcinoma in situ (LCIS). Math Biosci 2024; 380:109369. [PMID: 39694324 DOI: 10.1016/j.mbs.2024.109369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
Lobular carcinoma in situ (LCIS) is a precursor of invasive lobular carcinoma of the breast. LCIS cells lack cell-cell cohesion due to the loss of E-cadherin. LCIS cells grow in mammary lobules rather than in ducts. The etiology of this pattern, especially its dependence on cellular cohesion, is incompletely understood. We simulated passive intra-glandular scattering of carcinoma in situ (CIS) cells in an ultra-simplified hollow mold tissue replica (HMTR) and a discrete-time mathematical model featuring particles of variable sizes representing single cells (LCIS-like particles) or groups of cohesive carcinoma cells (DCIS-like particles). The HMTR features structures reminiscent of a mammary duct with associated lobules. The discrete mathematical model characterizes spatial redistribution over time and includes transition probabilities between ductal or lobular localizations. Redistribution of particles converged toward an equilibrium depending on particle size. Strikingly, equilibrium proportions depended on particle properties, which we also confirm in a continuous-time mathematical model that considers controlling lobular properties such as crowding. Particles of increasing size, representing CIS cells with proficient cohesion, showed increasingly higher equilibrium ductal proportions. Our investigations represent two conceptual abstractions implying a link between loss of cell-cell cohesion and lobular localization of LCIS, which provide a much-needed logical foundation for studying the connections between collective cell behavior and cancer development in breast tissues. In light of the findings from our simplified modeling approach, we discuss multiple avenues for near-future research that can address and evaluate the redistribution hypothesis mathematically and empirically.
Collapse
Affiliation(s)
- Matthias Christgen
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany.
| | - Rodrigo A Caetano
- Departamento de Física, Universidade Federal do Paraná, Curitiba, Brazil
| | - Michael Eisenburger
- Clinic for Prosthetic Dentistry and Biomedical Material Science, Hannover Medical School, Germany
| | - Arne Traulsen
- Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, August-Thienemann-Str. 2, Plön 24306, Germany
| | - Philipp M Altrock
- Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, August-Thienemann-Str. 2, Plön 24306, Germany.
| |
Collapse
|
11
|
Gentile F. The maximum size of cell-aggregates is determined by the competition between the strain energy and the binding energy of cells. Heliyon 2024; 10:e40560. [PMID: 39654728 PMCID: PMC11625300 DOI: 10.1016/j.heliyon.2024.e40560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
The development of tissues and organs is affected by how cells interact with each other to form aggregates. Such an interaction is in turn determined by several different factors, such as inter-cellular attractive forces, cell motility, and the strain energy of cells. Here, we have used mathematical modelling and numerical simulations to explore how the interplay between these factors can influence the formation and stability of 2D cell aggregates. Cell aggregates were created by incrementally accumulating cells over an initial seed. The binding energy density of these aggregates was determined using the harmonic approximation and was integrated into a probabilistic model to estimate the maximum cluster size, beyond which the aggregate becomes unstable and breaks into smaller fragments. Our simulations reveal that the ratio of strain energy to internal adhesive energy (U s / U b ) critically impacts cell aggregation; smaller ratios allow for larger cluster sizes. These findings have significant implications for tissue engineering, in-vitro modeling, the study of neurodegenerative diseases, and tissue regeneration, providing insights into how physical and biological characteristics of cells influence their aggregation and stability.
Collapse
Affiliation(s)
- Francesco Gentile
- Nanotechnology Research Center, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy
| |
Collapse
|
12
|
Dai L, Jin Y, Chai J, Yang J, Wang J, Chen M, Li L, Wang C, Yan G. Deficiency of DEK proto-oncogene alleviates allergic rhinitis by inhibiting RhoA/Ezrin-mediated mitochondrial fission. Animal Model Exp Med 2024. [PMID: 39668431 DOI: 10.1002/ame2.12523] [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: 08/22/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Allergic rhinitis (AR) is a kind of immune disease mediated by IgE. We are intrigued by the potential role of DEK proto-oncogene (DEK) in inflammation-related diseases. We investigated the effects and mechanisms of DEK in treating AR, aiming to identify potential new treatment targets for AR. METHODS The AR mouse model was induced by house dust mite (HDM) (1 mg/mL). HNEpCs stimulated by HDM (1 mg/mL) were pretreated for 24 h with or without DEK lentivirus. The effect of DEK knockout or knockdown on AR was evaluated in vitro and in vivo using western blotting, ELISA, flow cytometry, real-time quantitative PCR, immunohistochemistry, HE staining, PAS staining, Diff staining, and immunofluorescence. RESULTS After DEK knockdown, the inflammatory response of AR mice was reduced. In addition, DEK deletion mitigated nasal tissue damage and mitochondrial division. Our further studies showed that DEK deletion or inhibition led to the down-regulation of RhoA activity and decreased phosphorylation of Ezrin and Drp1 proteins, and inhibited mitochondrial division. Overall, DEK deficiency mitigated AR by down-regulating RhoA/Ezrin/Drp1 pathway activity. CONCLUSION DEK alleviates AR through RhoA/Ezrin/Drp1 signaling pathway, which provides a new perspective for developing improved therapies and understanding the pathogenesis of AR.
Collapse
Affiliation(s)
- Longzhu Dai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P. R. China
| | - Yongde Jin
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Otorhinolaryngology, Affiliated Hospital of Yanbian University, Yanji, P. R. China
| | - Jingmei Chai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Traditional Chinese Medicine, Yanbian University Medicine College, Yanji, P. R. China
| | - Jianing Yang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P. R. China
| | - Jiangang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P. R. China
| | - Mu Chen
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Otorhinolaryngology, Affiliated Hospital of Yanbian University, Yanji, P. R. China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P. R. China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P. R. China
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, P. R. China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P. R. China
| |
Collapse
|
13
|
Cai G, Rodgers NC, Liu AP. Unjamming Transition as a Paradigm for Biomechanical Control of Cancer Metastasis. Cytoskeleton (Hoboken) 2024. [PMID: 39633605 DOI: 10.1002/cm.21963] [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: 08/14/2024] [Revised: 10/27/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Tumor metastasis is a complex phenomenon that poses significant challenges to current cancer therapeutics. While the biochemical signaling involved in promoting motile phenotypes is well understood, the role of biomechanical interactions has recently begun to be incorporated into models of tumor cell migration. Specifically, we propose the unjamming transition, adapted from physical paradigms describing the behavior of granular materials, to better discern the transition toward an invasive phenotype. In this review, we introduce the jamming transition broadly and narrow our discussion to the different modes of 3D tumor cell migration that arise. Then we discuss the mechanical interactions between tumor cells and their neighbors, along with the interactions between tumor cells and the surrounding extracellular matrix. We center our discussion on the interactions that induce a motile state or unjamming transition in these contexts. By considering the interplay between biochemical and biomechanical signaling in tumor cell migration, we can advance our understanding of biomechanical control in cancer metastasis.
Collapse
Affiliation(s)
- Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole C Rodgers
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Allen P Liu
- Applied Physics Program, University of Michigan, Ann Arbor, Michigan, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Zhang P, Meng W, Wei L, Li Y, Xiao H, He Y, Yang F, Han X, Shu W. Rational design of a NIR fluorescent probe and its application in food detection of viscosity and biosystem imaging. Food Chem 2024; 460:140527. [PMID: 39121776 DOI: 10.1016/j.foodchem.2024.140527] [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: 06/06/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
Viscosity is one of the most important parameters of liquid foods and shows significant change during food spoilage. It is also an important component of the cell microenvironment and is closely associated with the development of liver injury. In this work, a viscosity-sensitive fluorescent probe named WZ-V based on the twisted intramolecular charge transfer (TICT) mechanism was successfully designed. WZ-V had a large Stokes shift, long wavelength emission, and the fluorescence intensity shows 290-fold enhancement in high viscosity. Probe WZ-V successfully detected viscosity changes caused by food thickeners, as well as in milk, orange juice, and lemonade spoilage processes. This provides a new tool for regulating the viscosity of liquid foods and monitoring viscosity changes during food spoilage. In addition, WZ-V has been successfully applied to image viscosity changes in liver injury, which provides an important reference for the study of liver diseases.
Collapse
Affiliation(s)
- Peng Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China; School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Wenshu Meng
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Liangchen Wei
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Yumeng Li
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Haibin Xiao
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255000, China
| | - Yongrui He
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Fengtang Yang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Wei Shu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China.
| |
Collapse
|
15
|
Zhao Q, Han B, Wang L, Wu J, Wang S, Ren Z, Wang S, Yang H, Carbone M, Dong C, Melino G, Chen WL, Jia W. AKR1B1-dependent fructose metabolism enhances malignancy of cancer cells. Cell Death Differ 2024; 31:1611-1624. [PMID: 39406918 PMCID: PMC11618507 DOI: 10.1038/s41418-024-01393-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/15/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024] Open
Abstract
Fructose metabolism has emerged as a significant contributor to cancer cell proliferation, yet the underlying mechanisms and sources of fructose for cancer cells remain incompletely understood. In this study, we demonstrate that cancer cells can convert glucose into fructose through a process called the AKR1B1-mediated polyol pathway. Inhibiting the endogenous production of fructose through AKR1B1 deletion dramatically suppressed glycolysis, resulting in reduced cancer cell migration, inhibited growth, and the induction of apoptosis and cell cycle arrest. Conversely, the acceleration of endogenous fructose through AKR1B1 overexpression has been shown to significantly enhance cancer cell proliferation and migration with increased S cell cycle progression. Our findings highlight the crucial role of endogenous fructose in cancer cell malignancy and support the need for further investigation into AKR1B1 as a potential cancer therapeutic target.
Collapse
Affiliation(s)
- Qing Zhao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Bing Han
- Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Lu Wang
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Jia Wu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Siliang Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhenxing Ren
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shouli Wang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haining Yang
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Michele Carbone
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Changsheng Dong
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy.
| | - Wen-Lian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
16
|
Chen P, Dong B, Yao W. Numerical simulation study of nanoparticle diffusion in gray matter. Comput Struct Biotechnol J 2024; 25:95-104. [PMID: 38974013 PMCID: PMC11225016 DOI: 10.1016/j.csbj.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose Nanomedicine-based approaches have shown great potential in the treatment of central nervous system diseases. However, the fate of nanoparticles (NPs) within the brain parenchyma has not received much attention. The complexity of the microstructure of the brain and the invisibility of NPs make it difficult to study NP transport within the grey matter. Moreover, regulation of NP delivery is not fully understood. Methods 2D interstitial system (ISS) models reflecting actual extracellular space (ECS) were constructed. A particle tracing model was used to simulate the diffusion of the NPs. The effect of NP size on NP diffusion was studied using numerical simulations. The diffusion of charged NPs was explored by comparing experimental and numerical simulation data, and the effect of cell membrane potential on the diffusion of charged NPs was further studied. Results The model was verified using previously published experimental data. Small NPs could diffuse efficiently into the ISS. The diffusion of charged NPs was hindered in the ISS. Changes in cell membrane potential had little effect on NP diffusion. Conclusion This study constructed 2D brain ISS models that reflected the actual ECS and simulated the diffusion of NPs within it. The study found that uncharged small NPs could effectively diffuse within the ISS and that the cell membrane potential had a limited effect on the diffusion of charged NPs. The model and findings of this study can aid the design of nanomedicines and nanocarriers for the diagnosis and treatment of brain diseases.
Collapse
Affiliation(s)
- Peiqian Chen
- Tongren Hospital, No. 1111, Xianxia Rd., Shanghai, China
- School of Medicine, Shanghai Jiao Tong University, No. 280, South Chongqing Rd., Shanghai, China
| | - Bing Dong
- School of Nuclear Science and Engineering, Shanghai Jiao Tong University, No. 800, Dongchuan Rd., Shanghai, China
| | - Weiwu Yao
- Tongren Hospital, No. 1111, Xianxia Rd., Shanghai, China
- School of Medicine, Shanghai Jiao Tong University, No. 280, South Chongqing Rd., Shanghai, China
| |
Collapse
|
17
|
Qianqian J, He W, Kaiguang Y, Baofeng Z, Zhen L, Yukui Z, Bo J, Lihua Z. 2D Nano-Photosensitizer Facilitates Proximity Labeling for Living Cells Surfaceome Deciphering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407240. [PMID: 39529546 DOI: 10.1002/smll.202407240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Photocatalytic proximity labeling has shown great promise for mapping the spatiotemporal dynamics of surfaceome. Although cell-surface targeting photosensitizers relying on antibodies, lipid molecules, and metabolic labeling have gained effects, the development of simpler and stable methods that avoid complex chemical synthesis and biosynthesis steps is still a huge challenge. Here, the study has introduced 2D nanomaterials with the ability of cell surface engineering to perform the in situ anchoring of photosensitizer on living cell surface. Photosensitizer can be stabilized on nanomaterials by coordination after one-step mixing, resulting in the nano-photosensitizer combining cell surface targeting ability and photosensitivity that allowing surface-specific proximity labeling. Nano-photosensitizer can be dispersed stably in aqueous solution, avoiding the defects of poor water solubility and aggregation of traditional organic photosensitizers. Singlet oxygen is generated locally under light irradiation, enabling spatiotemporally-resolved activating and labeling of cell surface proteome. Further application in the brain metastatic lung cancer has been found effective with numerous quantified differential cell surfaces proteins highly correlated with cancer metastasis and three potential players have been validated via immunoblotting and immunofluorescence, providing important insights for metastasis supported molecular mechanism.
Collapse
Affiliation(s)
- Jiang Qianqian
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wang He
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Kaiguang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhao Baofeng
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Liang Zhen
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhang Yukui
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Jiang Bo
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhang Lihua
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| |
Collapse
|
18
|
Xiao H, Gong X, Jordan SN, Liang Z, Mak M. Viscosity regulates cell spreading and cell-extracellular matrix interactions. FEBS J 2024. [PMID: 39529371 DOI: 10.1111/febs.17306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/16/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Fluid viscosity and osmolarity are among some of the underappreciated mechanical stimuli that cells can detect. Abnormal changes of multiple fluidic factors such as viscosity and osmolarity have been linked with diseases such as cystic fibrosis, cancer, and coronary heart disease. Changes in viscosity have been recently suggested as a regulator of cell locomotion. These novel studies focus on cell migration and spreading on glass substrates and through microchannels, and it remains a question whether viscosity impacts the cellular remodeling of extracellular matrices (ECMs). Here, we demonstrate that elevated viscosity induces cellular remodeling of collagen substrates and enhances cell spreading on ECM-mimetic substrates. Our results expand on recent work showing that viscosity induces increased cellular forces and demonstrates that viscosity can drive local ECM densification. Our data further show that microtubules, Ras-related C3 botulinum toxin substrate 1 (Rac1), actin-related protein 2/3 (Arp2/3) complex, Rho-associated protein kinase 1 (ROCK), and myosin are important regulators of viscosity-induced ECM remodeling. In the context of viscosity-induced cell spreading, cells cultured on glass and collagen substrates exhibit markedly different responses to pharmacological treatments, indicating that microtubules, Rac1, and Arp2/3 play distinct roles in regulating cellular spreading depending on the substrate. In addition, our results demonstrate that high osmotic pressures override viscosity-induced cell spreading by suppressing membrane ruffling. Our results demonstrate viscosity as a regulator of ECM remodeling and cell spreading in a fibrillar microenvironment. We also reveal a complex interplay between viscosity and osmolarity. We anticipate that our research can pave the way for future investigations into the crucial roles played by viscosity in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Seyma Nayir Jordan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Zixie Liang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
19
|
Yang Y, Yuan T, Panaitescu C, Li R, Wu K, Zhou Y, Pokrajac D, Dini D, Zhan W. Exploring tissue permeability of brain tumours in different grades: Insights from pore-scale fluid dynamics analysis. Acta Biomater 2024:S1742-7061(24)00656-1. [PMID: 39522625 DOI: 10.1016/j.actbio.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Interstitial fluid (ISF) flow is identified as an essential physiological process that plays an important role in the development and progression of brain tumours. However, the relationship between the permeability of the tumour tissue, a complex porous medium, and the interstitial fluid flow around the tumour cells at the microscale is not well understood. To shed light on this issue, and in the absence of experimental techniques that can provide direct measurements, we develop a computational model to predict the tissue permeability of brain tumours in different grades by analysing the ISF flow at the pore scale. The 3-D geometrical models of tissue extracellular spaces are digitally reconstructed for each grade tumour based on their morphological properties measured from microscopic images. The predictive accuracy of the framework is validated by experimental results reported in the literature. Our results indicate that high-grade brain tumours are less permeable despite their higher porosity, whereas necrotic areas of glioblastoma are more permeable than the viable tumour areas. This implies that tissue permeability is primarily governed by both tissue porosity and the deposition of hyaluronic acid (HA), a key component of the extracellular matrix, while the HA deposition can have a greater effect than macro-level porosity. Parametric studies show that tissue permeability falls exponentially with increasing HA concentration in all grades of brain tumours, and this can be captured using an empirically derived relationship in a quantitative manner. These findings provide an improved understanding of the hydraulic properties of brain tumours and their intrinsic links to tumour microstructure. This work can be used to reveal the intratumoural physiochemical processes that rely on fluid flow and offer a powerful tool to tune textured and porous biomaterials for desired transport properties. STATEMENT OF SIGNIFICANCE: Interstitial fluid flow in the extracellular space of brain tumours plays a crucial role in their progression, development, and response to drug treatments. However, the mechanisms of interstitial fluid transport around tumour cells and the characterization of these microscale transports at the tissue scale to meet clinical requirements are largely unknown. In the absence of advanced experimental techniques to capture these pore-scale transport phenomena, we have developed and validated a computational framework to successfully reveal these phenomena across all grades of brain tumours. For the first time, we have quantitatively determined the tissue permeability of all grades of brain tumours as a function of the concentration of hyaluronic acid, a key component of the extracellular matrix. This framework will enhance our ability to capture the intratumoural physicochemical processes in brain tumours and correlate them with tumour tissue-scale behaviours.
Collapse
Affiliation(s)
- Yi Yang
- School of Engineering, University of Aberdeen, Aberdeen AB24 3UE, UK.
| | - Tian Yuan
- Department of Mechanical Engineering, Imperial College London, London SW7 2AZ, UK
| | | | - Rui Li
- School of Engineering, University of Aberdeen, Aberdeen AB24 3UE, UK
| | - Kejian Wu
- School of Engineering, University of Aberdeen, Aberdeen AB24 3UE, UK
| | - Yingfang Zhou
- School of Engineering, University of Aberdeen, Aberdeen AB24 3UE, UK
| | - Dubravka Pokrajac
- School of Engineering, University of Aberdeen, Aberdeen AB24 3UE, UK
| | - Daniele Dini
- Department of Mechanical Engineering, Imperial College London, London SW7 2AZ, UK.
| | - Wenbo Zhan
- School of Engineering, University of Aberdeen, Aberdeen AB24 3UE, UK.
| |
Collapse
|
20
|
Huang Z, Xiao F, Wang Q, Zhang X, Shen Y, Deng Y, Shi P. BSC2 modulates AmB resistance via the maintenance of intracellular sodium/potassium ion homeostasis in Saccharomyces cerevisiae. Res Microbiol 2024; 175:104245. [PMID: 39245192 DOI: 10.1016/j.resmic.2024.104245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/13/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Previous studies on BSC2 have shown that it enhances yeast cell resistance to AmB via antioxidation and induces multidrug resistance by contributing to biofilm formation. Herein, we found that BSC2 overexpression could reverse the sensitivity of pmp3Δ to AmB and help the tested strains restore the intracellular sodium/potassium balance under exposure to AmB. Meanwhile, overexpression of the chitin gene CHS2 could simulate BSC2 to reverse the sensitivity of pmp3Δ and nha1Δ to high salt or AmB. However, BSC2 overexpression in flo11Δ failed to induce AmB resistance, form biofilms, and affect cell wall biogenesis, while CHS2 overexpression compensated the resistance of flo11Δ to AmB. Additionally, BSC2 levels were positively correlated with maintaining cell membrane integrity under exposure to AmB, CAS, or a combination of both. BSC2 overexpression in nha1Δ exhibited a similar function of CHS2, which can compensate for the sensitivity of the mutant to high salt. Altogether, the results demonstrate for the first time that BSC2 may promote ion equilibrium by strengthening cell walls and inhibiting membrane damage in a FLO path-dependent manner, thus enhancing the resistance of yeast cells to AmB. This study also reveals the possible mechanism of antifungal drugs CAS and AmB combined to inhibit fungi.
Collapse
Affiliation(s)
- Zhiwei Huang
- College of Biological Science and Medical Engineering, Donghua University, 2999 Renmin Road, Shanghai, 201620, China.
| | - Fulong Xiao
- College of Biological Science and Medical Engineering, Donghua University, 2999 Renmin Road, Shanghai, 201620, China
| | - Qiao Wang
- College of Biological Science and Medical Engineering, Donghua University, 2999 Renmin Road, Shanghai, 201620, China
| | - Xiaojuan Zhang
- College of Biological Science and Medical Engineering, Donghua University, 2999 Renmin Road, Shanghai, 201620, China
| | - Yuhu Shen
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Qinghai Provincial Key Laboratory of Crop Molecular Breeding, Northwest Institute of Plateau Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Xining, 810008, China; Laboratory for Research and Utilization of Qinghai Tibet Plateau Germplasm Resources, Academy of Agriculture and Forestry Sciences of Qinghai University (Qinghai Academy of Agriculture and Forestry Sciences), Xining, 810016, China
| | - Yunxia Deng
- College of Biological Science and Medical Engineering, Donghua University, 2999 Renmin Road, Shanghai, 201620, China
| | - Ping Shi
- Laboratory for Research and Utilization of Qinghai Tibet Plateau Germplasm Resources, Academy of Agriculture and Forestry Sciences of Qinghai University (Qinghai Academy of Agriculture and Forestry Sciences), Xining, 810016, China; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| |
Collapse
|
21
|
Chang Z, Zhou Y, Dong L, Qiao LR, Yang H, Xu GK. Deciphering the complex mechanics of atherosclerotic plaques: A hybrid hierarchical theory-microrheology approach. Acta Biomater 2024; 189:399-412. [PMID: 39307259 DOI: 10.1016/j.actbio.2024.09.029] [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: 03/19/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024]
Abstract
Understanding the viscoelastic properties of atherosclerotic plaques at rupture-prone scales is crucial for assessing their vulnerability. Here, we develop a Hybrid Hierarchical theory-Microrheology (HHM) approach, enabling the analysis of multiscale mechanical variations and distribution changes in regional tissue viscoelasticity within plaques across different spatial scales. We disclose a universal two-stage power-law rheology in plaques, characterized by distinct power-law exponents (αshort and αlong), which serve as mechanical indexes for plaque components and assessing mechanical gradients. We further propose a self-similar hierarchical theory that effectively delineates plaque heterogeneity from the cytoplasm, cell, to tissue levels. Moreover, our proposed multi-layer perceptron model addresses the viscoelastic heterogeneity and gradients within plaques, offering a promising diagnostic strategy for identifying unstable plaques. These findings not only advance our understanding of plaque mechanics but also pave the way for innovative diagnostic approaches in cardiovascular disease management. STATEMENT OF SIGNIFICANCE: Our study pioneers a Hybrid Hierarchical theory-Microrheology (HHM) approach to dissect the intricate viscoelasticity of atherosclerotic plaques, focusing on distinct components including cap fibrosis, lipid pools, and intimal fibrosis. We unveil a universal two-stage power-law rheology capturing mechanical variations across plaque structures. The proposed hierarchical model adeptly captures viscoelasticity changes from cytoplasm, cell to tissue levels. Based on the newly proposed markers, we further develop a machine learning (ML) diagnostic model that sets precise criteria for evaluating plaque components and heterogeneity. This work not only reveals the comprehensive mechanical heterogeneity within plaques but also introduces a mechanical marker-based ML strategy for assessing plaque conditions, offering a significant leap towards understanding and diagnosing atherosclerotic risks.
Collapse
Affiliation(s)
- Zhuo Chang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, China
| | - Le Dong
- School of Artificial Intelligence, Xidian University, Xi'an 710071, China
| | - Lin-Ru Qiao
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, China.
| | - Guang-Kui Xu
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
22
|
Yang Y, Chen S, Zhang L, Zhang G, Liu Y, Li Y, Zou L, Meng L, Tian Y, Dai L, Xiong M, Pan L, Xiong J, Chen L, Hou H, Yu Z, Zhang Z. The PM20D1-NADA pathway protects against Parkinson's disease. Cell Death Differ 2024; 31:1545-1560. [PMID: 39174646 PMCID: PMC11519464 DOI: 10.1038/s41418-024-01356-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024] Open
Abstract
Parkinson's disease (PD) is characterized by the selective loss of dopaminergic neurons in the substantia nigra and the accumulation of α-synuclein (α-Syn) aggregates. However, the molecular mechanisms regulating α-Syn aggregation and neuronal degeneration remain poorly understood. The peptidase M20 domain containing 1 (PM20D1) gene lies within the PARK16 locus genetically linked to PD. Single nucleotide polymorphisms regulating PM20D1 expression are associated with changed risk of PD. Dopamine (DA) metabolism and DA metabolites have been reported to regulate α-Syn pathology. Here we report that PM20D1 catalyzes the conversion of DA to N-arachidonoyl dopamine (NADA), which interacts with α-Syn and inhibits its aggregation. Simultaneously, NADA competes with α-Syn fibrils to regulate TRPV4-mediated calcium influx and downstream phosphatases, thus alleviating α-Syn phosphorylation. The expression of PM20D1 decreases during aging. Overexpression of PM20D1 or the administration of NADA in a mouse model of synucleinopathy alleviated α-Syn pathology, dopaminergic neurodegeneration, and motor impairments. These observations support the protective effect of the PM20D1-NADA pathway against the progression of α-Syn pathology in PD.
Collapse
Affiliation(s)
- Yunying Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Sichun Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yan Liu
- Department of Nursing, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiming Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Zou
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Tian
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lina Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liam Chen
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hua Hou
- Department of Polymer Science, College of Chemistry and Molecular Sciences of Wuhan University, Wuhan, 430060, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
23
|
Lan BQ, Wang YJ, Yu SX, Liu W, Liu YJ. Physical effects of 3-D microenvironments on confined cell behaviors. Am J Physiol Cell Physiol 2024; 327:C1192-C1201. [PMID: 39246142 DOI: 10.1152/ajpcell.00288.2024] [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: 04/30/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Cell migration is a fundamental and functional cellular process, influenced by a complex microenvironment consisting of different cells and extracellular matrix. Recent research has highlighted that, besides biochemical cues from the microenvironment, physical cues can also greatly alter cellular behavior. However, due to the complexity of the microenvironment, little is known about how the physical interactions between migrating cells and surrounding microenvironment instructs cell movement. Here, we explore various examples of three-dimensional microenvironment reconstruction models in vitro and describe how the physical interplay between migrating cells and the neighboring microenvironment controls cell behavior. Understanding this mechanical cooperation will provide key insights into organ development, regeneration, and tumor metastasis.
Collapse
Affiliation(s)
- Bao-Qiong Lan
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| |
Collapse
|
24
|
Chen X, Xu S, Chu B, Guo J, Zhang H, Sun S, Song L, Feng XQ. Applying Spatiotemporal Modeling of Cell Dynamics to Accelerate Drug Development. ACS NANO 2024; 18:29311-29336. [PMID: 39420743 DOI: 10.1021/acsnano.4c12599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cells act as physical computational programs that utilize input signals to orchestrate molecule-level protein-protein interactions (PPIs), generating and responding to forces, ultimately shaping all of the physiological and pathophysiological behaviors. Genome editing and molecule drugs targeting PPIs hold great promise for the treatments of diseases. Linking genes and molecular drugs with protein-performed cellular behaviors is a key yet challenging issue due to the wide range of spatial and temporal scales involved. Building predictive spatiotemporal modeling systems that can describe the dynamic behaviors of cells intervened by genome editing and molecular drugs at the intersection of biology, chemistry, physics, and computer science will greatly accelerate pharmaceutical advances. Here, we review the mechanical roles of cytoskeletal proteins in orchestrating cellular behaviors alongside significant advancements in biophysical modeling while also addressing the limitations in these models. Then, by integrating generative artificial intelligence (AI) with spatiotemporal multiscale biophysical modeling, we propose a computational pipeline for developing virtual cells, which can simulate and evaluate the therapeutic effects of drugs and genome editing technologies on various cell dynamic behaviors and could have broad biomedical applications. Such virtual cell modeling systems might revolutionize modern biomedical engineering by moving most of the painstaking wet-laboratory effort to computer simulations, substantially saving time and alleviating the financial burden for pharmaceutical industries.
Collapse
Affiliation(s)
- Xindong Chen
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
- BioMap, Beijing 100144, China
| | - Shihao Xu
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Bizhu Chu
- School of Pharmacy, Shenzhen University, Shenzhen 518055, China
- Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jing Guo
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Huikai Zhang
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Shuyi Sun
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Le Song
- BioMap, Beijing 100144, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
25
|
Lu C, Zeng T, Wang M, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Influence of viscosity on adipogenic and osteogenic differentiation of mesenchymal stem cells during 2D culture. Biomater Sci 2024; 12:5598-5609. [PMID: 39327896 DOI: 10.1039/d4bm00710g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Accumulatively, cellular behaviours triggered by biochemical cues have been widely explored and the focus of research is gradually shifting to biophysical cues. Compared to physical parameters such as stiffness, substrate morphology and viscoelasticity, the influence of viscosity on cellular behaviours is relatively unexplored and overlooked. Thus, in this study, the influence of viscosity on the adipogenic and osteogenic differentiation of human mesenchymal stem cells (hMSCs) was investigated by adjusting the viscosity of the culture medium. Viscosity exhibited different effects on adipogenic and osteogenic differentiation of hMSCs during two-dimensional (2D) culture. High viscosity facilitated osteogenic while inhibiting adipogenic differentiation. During adipogenic differentiation, the effect of viscosity on cell proliferation was negligible. However, during osteogenic differentiation, high viscosity decreased cell proliferation. The different influence of viscosity could be explained by the activation of mechanotransduction regulators of Yes-associated protein (YAP) and β-catenin. High viscosity could promote YAP and β-catenin nuclear translocation during osteogenic differentiation, which was responsible for the increased osteogenesis. High viscosity inhibited adipogenesis through promoting YAP nuclear translocation. This study could broaden the understanding of how viscosity can affect stem cell differentiation during 2D culture, which is valuable for tissue engineering.
Collapse
Affiliation(s)
- Chengyu Lu
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Tianjiao Zeng
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Man Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Toru Yoshitomi
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
| | - Yingnan Yang
- Graduate School of Life and Environment Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki 305-0044, Japan.
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
26
|
Yan G, Zhou J, Yin J, Gao D, Zhong X, Deng X, Kang H, Sun A. Membrane Ruffles: Composition, Function, Formation and Visualization. Int J Mol Sci 2024; 25:10971. [PMID: 39456754 PMCID: PMC11507850 DOI: 10.3390/ijms252010971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Membrane ruffles are cell actin-based membrane protrusions that have distinct structural characteristics. Linear ruffles with columnar spike-like and veil-like structures assemble at the leading edge of cell membranes. Circular dorsal ruffles (CDRs) have no supporting columnar structures but their veil-like structures, connecting from end to end, present an enclosed ring-shaped circular outline. Membrane ruffles are involved in multiple cell functions such as cell motility, macropinocytosis, receptor internalization, fluid viscosity sensing in a two-dimensional culture environment, and protecting cells from death in response to physiologically compressive loads. Herein, we review the state-of-the-art knowledge on membrane ruffle structure and function, the growth factor-induced membrane ruffling process, and the growth factor-independent ruffling mode triggered by calcium and other stimulating factors, together with the respective underlying mechanisms. We also summarize the inhibitors used in ruffle formation studies and their specificity. In the last part, an overview is given of the various techniques in which the membrane ruffles have been visualized up to now.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongyan Kang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (G.Y.); (J.Z.); (J.Y.); (D.G.); (X.Z.); (X.D.)
| | - Anqiang Sun
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (G.Y.); (J.Z.); (J.Y.); (D.G.); (X.Z.); (X.D.)
| |
Collapse
|
27
|
Caruso AP, Logue JS. The biophysics of cell motility through mechanochemically challenging environments. Curr Opin Cell Biol 2024; 90:102404. [PMID: 39053178 PMCID: PMC11392632 DOI: 10.1016/j.ceb.2024.102404] [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: 04/30/2024] [Revised: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024]
Abstract
Challenging mechanochemical environments (i.e., with varied mechanical and adhesive properties) are now known to induce a wide range of adaptive phenomena in motile cells. For instance, confinement and low adhesion may trigger a phenotypic transition to fast amoeboid (leader bleb-based) migration. The molecular mechanisms that underly these phenomena are beginning to be understood. Due to its size, the mechanical properties of the nucleus have been shown to limit and facilitate cell migration. Additionally, the activity of various transient receptor potential (TRP) channels is now known to be integral to cell migration in response to a multitude of biophysical stimuli. How cells integrate signals from the nucleus and plasma membrane, however, is unclear. The development of therapeutics that suppress cancer or enhance immune cell migration for immuno-oncology applications, etc., will require additional work to completely understand the molecular mechanisms that enable cells to navigate mechanochemically challenging environments.
Collapse
Affiliation(s)
- Alexa P Caruso
- Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA
| | - Jeremy S Logue
- Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, USA.
| |
Collapse
|
28
|
Mistriotis P, Wisniewski EO, Si BR, Kalab P, Konstantopoulos K. Coordinated in confined migration: crosstalk between the nucleus and ion channel-mediated mechanosensation. Trends Cell Biol 2024; 34:809-825. [PMID: 38290913 PMCID: PMC11284253 DOI: 10.1016/j.tcb.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Cell surface and intracellular mechanosensors enable cells to perceive different geometric, topographical, and physical cues. Mechanosensitive ion channels (MICs) localized at the cell surface and on the nuclear envelope (NE) are among the first to sense and transduce these signals. Beyond compartmentalizing the genome of the cell and its transcription, the nucleus also serves as a mechanical gauge of different physical and topographical features of the tissue microenvironment. In this review, we delve into the intricate mechanisms by which the nucleus and different ion channels regulate cell migration in confinement. We review evidence suggesting an interplay between macromolecular nuclear-cytoplasmic transport (NCT) and ionic transport across the cell membrane during confined migration. We also discuss the roles of the nucleus and ion channel-mediated mechanosensation, whether acting independently or in tandem, in orchestrating migratory mechanoresponses. Understanding nuclear and ion channel sensing, and their crosstalk, is critical to advancing our knowledge of cell migration in health and disease.
Collapse
Affiliation(s)
| | - Emily O Wisniewski
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bishwa R Si
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
29
|
Vázquez-Aristizabal P, Henriksen-Lacey M, García-Astrain C, Jimenez de Aberasturi D, Langer J, Epelde C, Litti L, Liz-Marzán LM, Izeta A. Biofabrication and Monitoring of a 3D Printed Skin Model for Melanoma. Adv Healthc Mater 2024; 13:e2401136. [PMID: 38992996 DOI: 10.1002/adhm.202401136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/27/2024] [Indexed: 07/13/2024]
Abstract
There is an unmet need for in vitro cancer models that emulate the complexity of human tissues. 3D-printed solid tumor micromodels based on decellularized extracellular matrices (dECMs) recreate the biomolecule-rich matrix of native tissue. Herein a 3D in vitro metastatic melanoma model that is amenable for drug screening purposes and recapitulates features of both the tumor and the skin microenvironment is described. Epidermal, basement membrane, and dermal biocompatible inks are prepared by means of combined chemical, mechanical, and enzymatic processes. Bioink printability is confirmed by rheological assessment and bioprinting, and bioinks are subsequently combined with melanoma cells and dermal fibroblasts to build complex 3D melanoma models. Cells are tracked by confocal microscopy and surface-enhanced Raman spectroscopy (SERS) mapping. Printed dECMs and cell tracking allow modeling of the initial steps of metastatic disease, and may be used to better understand melanoma cell behavior and response to drugs.
Collapse
Affiliation(s)
- Paula Vázquez-Aristizabal
- Stem Cells and Aging Group, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain s/n, Donostia-San Sebastián, 20014, Spain
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
| | - Malou Henriksen-Lacey
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
| | - Clara García-Astrain
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
| | - Dorleta Jimenez de Aberasturi
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, 48009, Spain
| | - Judith Langer
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
| | - Claudia Epelde
- Obstetrics and Gynaecology Service, Donostia University Hospital, Paseo Dr. Begiristain s/n, Donostia-San Sebastián, 20014, Spain
| | - Lucio Litti
- Department of Chemical Sciences, University of Padova, Via Marzolo, 1, Padova, 35131, Italy
| | - Luis M Liz-Marzán
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, Donostia-San Sebastián, 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián, 20014, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, 48009, Spain
- Cinbio, Universidade de Vigo, Campus Universitario, Vigo, 36310, Spain
| | - Ander Izeta
- Stem Cells and Aging Group, Biogipuzkoa Health Research Institute, Paseo Dr. Begiristain s/n, Donostia-San Sebastián, 20014, Spain
- School of Engineering, Tecnun-University of Navarra, Donostia-San Sebastián, 20009, Spain
| |
Collapse
|
30
|
Arpinati L, Carradori G, Scherz-Shouval R. CAF-induced physical constraints controlling T cell state and localization in solid tumours. Nat Rev Cancer 2024; 24:676-693. [PMID: 39251836 DOI: 10.1038/s41568-024-00740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Solid tumours comprise cancer cells that engage in continuous interactions with non-malignant cells and with acellular components, forming the tumour microenvironment (TME). The TME has crucial and diverse roles in tumour progression and metastasis, and substantial efforts have been dedicated into understanding the functions of different cell types within the TME. These efforts highlighted the importance of non-cell-autonomous signalling in cancer, mediating interactions between the cancer cells, the immune microenvironment and the non-immune stroma. Much of this non-cell-autonomous signalling is mediated through acellular components of the TME, known as the extracellular matrix (ECM), and controlled by the cells that secrete and remodel the ECM - the cancer-associated fibroblasts (CAFs). In this Review, we delve into the complex crosstalk among cancer cells, CAFs and immune cells, highlighting the effects of CAF-induced ECM remodelling on T cell functions and offering insights into the potential of targeting ECM components to improve cancer therapies.
Collapse
Affiliation(s)
- Ludovica Arpinati
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Carradori
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
31
|
Wang X, Li X, Liu Z, Meng Y, Fan X, Wang H, Nie J, Xue B. A golgi targeting viscosity rotor for cancer diagnosis in living cells and tissues. Talanta 2024; 278:126497. [PMID: 38955104 DOI: 10.1016/j.talanta.2024.126497] [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: 04/18/2024] [Revised: 06/15/2024] [Accepted: 06/29/2024] [Indexed: 07/04/2024]
Abstract
Unveiling the intricate relationship between cancer and Golgi viscosity remains an arduous endeavor, primarily due to the lack of Golgi-specific fluorescent probes tailored for viscosity measurement. Considering this formidable obstacle, we have triumphed over the challenge by devising a bespoke Golgi-specific viscosity probe, aptly named GOL-V. This ingenious innovation comprises the viscosity rotor BODIPY intricately tethered to the Golgi-targeting moiety benzsulfamide. GOL-V exhibits remarkable sensitivity to fluctuations in viscosity, the fluorescence intensity of GOL-V increased 114-fold when the viscosity value was increased from 2.63 to 937.28 cP. Owing to its remarkable capacity to suppress the TICT state under conditions of heightened viscosity. Moreover, its efficacy in sensitively monitoring Golgi viscosity alterations within living cells is also very significant. Astonishingly, our endeavors have culminated in not only the visualization of Golgi viscosity at the cellular and tissue levels but also in the clinical tissue samples procured from cancer patients. Harnessing the prowess of GOL-V, we have successfully demonstrated that Golgi viscosity could serve as a discerning marker for detecting the presence of cancer. The convergence of these exceptional attributes firmly establishes GOL-V as an immensely potent instrument, holding immense potential in the realm of cancer diagnosis.
Collapse
Affiliation(s)
- Xiaodong Wang
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, Taiyuan, 030031, PR China.
| | - Xiaoping Li
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, Taiyuan, 030031, PR China
| | - Zonghui Liu
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, Taiyuan, 030031, PR China
| | - Yating Meng
- Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xiaofang Fan
- Department of Pathology, Shanxi Provincial People's Hospital, Taiyuan, 030012, PR China
| | - Hui Wang
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, Taiyuan, 030031, PR China
| | - Jisheng Nie
- Shanxi Medical University, Taiyuan, 030001, PR China.
| | - Bingchun Xue
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, Taiyuan, 030031, PR China.
| |
Collapse
|
32
|
Gonçalves RC, Oliveira MB, Mano JF. Exploring the potential of all-aqueous immiscible systems for preparing complex biomaterials and cellular constructs. MATERIALS HORIZONS 2024; 11:4573-4599. [PMID: 39010747 DOI: 10.1039/d4mh00431k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
All-aqueous immiscible systems derived from liquid-liquid phase separation of incompatible hydrophilic agents such as polymers and salts have found increasing interest in the biomedical and tissue engineering fields in the last few years. The unique characteristics of aqueous interfaces, namely their low interfacial tension and elevated permeability, as well as the non-toxic environment and high water content of the immiscible phases, confer to these systems optimal qualities for the development of biomaterials such as hydrogels and soft membranes, as well as for the preparation of in vitro tissues derived from cellular assembly. Here, we overview the main properties of these systems and present a critical review of recent strategies that have been used for the development of biomaterials with increased levels of complexity using all-aqueous immiscible phases and interfaces, and their potential as cell-confining environments for micropatterning approaches and the bioengineering of cell-rich structures. Importantly, due to the relatively recent emergence of these areas, several key design considerations are presented, in order to guide researchers in the field. Finally, the main present challenges, future directions, and adaptability to develop advanced materials with increased biomimicry and new potential applications are briefly evaluated.
Collapse
Affiliation(s)
- Raquel C Gonçalves
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
33
|
Zhang F, Mehta H, Choudhary HH, Islam R, Hanafy KA. TRPV4 Channel in Neurological Disease: from Molecular Mechanisms to Therapeutic Potential. Mol Neurobiol 2024:10.1007/s12035-024-04518-5. [PMID: 39333347 DOI: 10.1007/s12035-024-04518-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) is a non-selective cation channel with pivotal roles in various physiological processes, including osmosensitivity, mechanosensation, neuronal development, vascular tone regulation, and bone homeostasis in human bodies. Recent studies have made significant progress in understanding the structure and functional role of TRPV4, shedding light on its involvement in pathological processes, particularly in the realm of neurological diseases. Here, we aim to provide a comprehensive exploration of the multifaceted contributions of TRPV4 to neurological diseases, spanning its intricate molecular mechanisms to its potential as a target for therapeutic interventions. We delve into the structural and functional attributes of TRPV4, scrutinize its expression profile, and elucidate the possible mechanisms through which it participates in the pathogenesis of neurological disorders. Furthermore, we discussed recent years' progress in therapeutic strategies aimed at harnessing TRPV4 for the treatment of these diseases. These insights will provide a basis for understanding and designing modality-specific pharmacological agents to treat TRPV4-associated disorders.
Collapse
Affiliation(s)
- Feng Zhang
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hritik Mehta
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hadi Hasan Choudhary
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Rezwanul Islam
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Khalid A Hanafy
- Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper University Health Care, Camden, NJ, USA.
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper Neurological Institute Center for Neuroinflammation, Cooper Medical School at Rowan University, Camden, NJ, USA.
| |
Collapse
|
34
|
Amiri F, Akinpelu AA, Keith WC, Hemmati F, Vaghasiya RS, Bowen D, Waliagha RS, Wang C, Chen P, Mitra AK, Li Y, Mistriotis P. Confinement controls the directional cell responses to fluid forces. Cell Rep 2024; 43:114692. [PMID: 39207902 PMCID: PMC11495937 DOI: 10.1016/j.celrep.2024.114692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/18/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Our understanding of how fluid forces influence cell migration in confining environments remains limited. By integrating microfluidics with live-cell imaging, we demonstrate that cells in tightly-but not moderately-confined spaces reverse direction and move upstream upon exposure to fluid forces. This fluid force-induced directional change occurs less frequently when cells display diminished mechanosensitivity, experience elevated hydraulic resistance, or sense a chemical gradient. Cell reversal requires actin polymerization to the new cell front, as shown mathematically and experimentally. Actin polymerization is necessary for the fluid force-induced activation of NHE1, which cooperates with calcium to induce upstream migration. Calcium levels increase downstream, mirroring the subcellular distribution of myosin IIA, whose activation enhances upstream migration. Reduced lamin A/C levels promote downstream migration of metastatic tumor cells by preventing cell polarity establishment and intracellular calcium rise. This mechanism could allow cancer cells to evade high-pressure environments, such as the primary tumor.
Collapse
Affiliation(s)
- Farshad Amiri
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Ayuba A Akinpelu
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - William C Keith
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Farnaz Hemmati
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Ravi S Vaghasiya
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Dylan Bowen
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Razan S Waliagha
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Chuanyu Wang
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Amit K Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; UAB O'Neal Comprehensive Cancer, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35233, USA
| | - Yizeng Li
- Department of Biomedical Engineering, Binghamton University, SUNY, Binghamton, NY 13902, USA
| | | |
Collapse
|
35
|
Ouyang P, Cheng B, He X, Lou J, Li X, Guo H, Xu F. Navigating the biophysical landscape: how physical cues steer the journey of bone metastatic tumor cells. Trends Cancer 2024; 10:792-808. [PMID: 39127608 DOI: 10.1016/j.trecan.2024.07.003] [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: 03/29/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
Many tumors prefer to metastasize to bone, but the underlying mechanisms remain elusive. The human skeletal system has unique physical properties, that are distinct from other organs, which play a key role in directing the behavior of tumor cells within bone. Understanding the physical journey of tumor cells within bone is crucial. In this review we discuss bone metastasis in the context of how physical cues in the bone vasculature and bone marrow niche regulate the fate of tumor cells. Our objective is to inspire innovative diagnostic and therapeutic approaches for bone metastasis from a mechanobiological perspective.
Collapse
Affiliation(s)
- Pengrong Ouyang
- Department of Orthopedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China; TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, P.R. China
| | - Xijing He
- Department of Orthopedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Xi'an International Medical Center Hospital, Xi'an 710061, P.R. China.
| | - Jiatao Lou
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China.
| | - Xiaokang Li
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, P.R. China.
| | - Hui Guo
- Department of Medical Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China.
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China.
| |
Collapse
|
36
|
Fister AM, Horn A, Lasarev MR, Huttenlocher A. Damage-induced basal epithelial cell migration modulates the spatial organization of redox signaling and sensory neuron regeneration. eLife 2024; 13:RP94995. [PMID: 39207919 PMCID: PMC11361710 DOI: 10.7554/elife.94995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Epithelial damage leads to early reactive oxygen species (ROS) signaling, which regulates sensory neuron regeneration and tissue repair. How the initial type of tissue injury influences early damage signaling and regenerative growth of sensory axons remains unclear. Previously we reported that thermal injury triggers distinct early tissue responses in larval zebrafish. Here, we found that thermal but not mechanical injury impairs sensory axon regeneration and function. Real-time imaging revealed an immediate tissue response to thermal injury characterized by the rapid Arp2/3-dependent migration of keratinocytes, which was associated with tissue scale ROS production and sustained sensory axon damage. Isotonic treatment was sufficient to limit keratinocyte movement, spatially restrict ROS production, and rescue sensory neuron function. These results suggest that early keratinocyte dynamics regulate the spatial and temporal pattern of long-term signaling in the wound microenvironment during tissue repair.
Collapse
Affiliation(s)
- Alexandra M Fister
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Adam Horn
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
| | - Michael R Lasarev
- Department of Biostatistics and Medical Informatics, University of Wisconsin-MadisonMadisonUnited States
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-MadisonMadisonUnited States
- Department of Pediatrics, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
37
|
Abdul Halim MS, Dyson JM, Gong MM, O'Bryan MK, Nosrati R. Fallopian tube rheology regulates epithelial cell differentiation and function to enhance cilia formation and coordination. Nat Commun 2024; 15:7411. [PMID: 39198453 PMCID: PMC11358425 DOI: 10.1038/s41467-024-51481-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
The rheological properties of the extracellular fluid in the female reproductive tract vary spatiotemporally, however, the effect on the behaviour of epithelial cells that line the tract is unexplored. Here, we reveal that epithelial cells respond to the elevated viscosity of culture media by modulating their development and functionality to enhance cilia formation and coordination. Specifically, ciliation increases by 4-fold and cilia beating frequency decreases by 30% when cells are cultured at 100 mPa·s. Further, cilia manifest a coordinated beating pattern that can facilitate the formation of metachronal waves. At the cellular level, viscous loading activates the TRPV4 channel in the epithelial cells to increase intracellular Ca2+, subsequently decreasing the mitochondrial membrane potential level for ATP production to maintain cell viability and function. Our findings provide additional insights into the role of elevated tubal fluid viscosity in promoting ciliation and coordinating their beating-a potential mechanism to facilitate the transport of egg and embryo, suggesting possible therapeutic opportunities for infertility treatment.
Collapse
Affiliation(s)
- Melati S Abdul Halim
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia
| | - Jennifer M Dyson
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Max M Gong
- Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Molecular Science and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Reza Nosrati
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
38
|
Torra J, Campelo F, Garcia-Parajo MF. Tensing Flipper: Photosensitized Manipulation of Membrane Tension, Lipid Phase Separation, and Raft Protein Sorting in Biological Membranes. J Am Chem Soc 2024; 146:24114-24124. [PMID: 39162019 PMCID: PMC11363133 DOI: 10.1021/jacs.4c08580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
The lateral organization of proteins and lipids in the plasma membrane is fundamental to regulating a wide range of cellular processes. Compartmentalized ordered membrane domains enriched with specific lipids, often termed lipid rafts, have been shown to modulate the physicochemical and mechanical properties of membranes and to drive protein sorting. Novel methods and tools enabling the visualization, characterization, and/or manipulation of membrane compartmentalization are crucial to link the properties of the membrane with cell functions. Flipper, a commercially available fluorescent membrane tension probe, has become a reference tool for quantitative membrane tension studies in living cells. Here, we report on a so far unidentified property of Flipper, namely, its ability to photosensitize singlet oxygen (1O2) under blue light when embedded into lipid membranes. This in turn results in the production of lipid hydroperoxides that increase membrane tension and trigger phase separation. In biological membranes, the photoinduced segregated domains retain the sorting ability of intact phase-separated membranes, directing raft and nonraft proteins into ordered and disordered regions, respectively, in contrast to radical-based photo-oxidation reactions that disrupt raft protein partitioning. The dual tension reporting and photosensitizing abilities of Flipper enable simultaneous visualization and manipulation of the mechanical properties and lateral organization of membranes, providing a powerful tool to optically control lipid raft formation and to explore the interplay between membrane biophysics and cell function.
Collapse
Affiliation(s)
- Joaquim Torra
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Barcelona 08860, Spain
| | - Felix Campelo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Barcelona 08860, Spain
| | - Maria F Garcia-Parajo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Barcelona 08860, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| |
Collapse
|
39
|
Jia X, Shen D, Deng J, Wang K, Wang X, Guo Y, Sun L, Jin H, Xia Q, Feng H, Jing B, Sun J, Wan W, Liu Y, Li M. Isophorone-based crystallization-induced-emission sensors detect proteome aggregation in live cells and tissues with breast cancer. Anal Chim Acta 2024; 1317:342916. [PMID: 39030013 DOI: 10.1016/j.aca.2024.342916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Protein misfolding and aggregation can lead to various diseases. Recent studies have shed light on the aggregated protein in breast cancer pathology, which suggests that it is crucial to design chemical sensors that visualize protein aggregates in breast cancer, especially in clinical patient-derived samples. However, most reported sensors are constrained in cultured cell lines. RESULTS In this work, we present the development of two isophorone-based crystallization-induced-emission fluorophores for detecting proteome aggregation in breast cancer cell line and tissues biopsied from diseased patients, designated as A1 and A2. These probes exhibited viscosity sensitivity and recovered their fluorescence strongly at crystalline state. Moreover, A1 and A2 exhibit selective binding capacity and strong fluorescence for various aggregated proteins. Utilizing these probes, we detect protein aggregation in stressed breast cancer cells, xenograft mouse model of human breast cancer and clinical patient-derived samples. Notably, the fluorescence intensity of both probes light up in tumor tissues. SIGNIFICANCE The synthesized isophorone-based crystallization-induced-emission fluorophores, A1 and A2, enable sensitive detection of protein aggregation in breast cancer cells and tissues. In the future, aggregated proteins are expected to become indicators for early diagnosis and clinical disease monitoring of breast cancer.
Collapse
Affiliation(s)
- Xiaomeng Jia
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China
| | - Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China
| | - Jintai Deng
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China
| | - Kainan Wang
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China
| | - Xueqing Wang
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China
| | - Yijin Guo
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China
| | - Lu Sun
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China
| | - Hao Jin
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China
| | - Qiuxuan Xia
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Huan Feng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Biao Jing
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China
| | - Jialu Sun
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China
| | - Wang Wan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China
| | - Yu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China.
| | - Man Li
- The Second Hospital of Dalian Medical University, Dalian, 116023, PR China.
| |
Collapse
|
40
|
Han X, Liu J, Zhang Y, Tse E, Yu Q, Lu Y, Ma Y, Zheng L. Increasing the tumour targeting of antitumour drugs through anlotinib-mediated modulation of the extracellular matrix and the RhoA/ROCK signalling pathway. J Pharm Anal 2024; 14:100984. [PMID: 39258171 PMCID: PMC11386283 DOI: 10.1016/j.jpha.2024.100984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 09/12/2024] Open
Abstract
Anlotinib has strong antiangiogenic effects and leads to vessel normalization. However, the "window period" characteristic in regulating vessel normalization by anlotinib cannot fully explain the long-term survival benefits achieved through combining it with other drugs. In this study, through RNA sequencing (RNA-seq) and label-free quantitative proteomics analysis, we discovered that anlotinib regulated the expression of components of the extracellular matrix (ECM), leading to a significant reduction in ECM stiffness. Our bioinformatic analysis revealed a potential positive relationship between the ECM pathway and gefitinib resistance, poor treatment outcomes for programmed death 1 (PD-1) targeting, and unfavourable prognosis following chemotherapy in lung cancer patients. We administered anlotinib in combination with these antitumour drugs and visualized their distribution using fluorescent labelling in various tumour types. Notably, our results demonstrated that anlotinib prolonged the retention time and distribution of antitumour drugs at the tumour site. Moreover, the combination therapy induced notable loosening of the tumour tissue structure. This reduction was associated with decreased interstitial fluid pressure and tumour solid pressure. Additionally, we observed that anlotinib effectively suppressed the Ras homologue family member A (RhoA)/Rho-associated protein kinase (ROCK) signalling pathway. These findings suggest that, in addition to its antiangiogenic and vessel normalization effects, anlotinib can increase the distribution and retention of antitumour drugs in tumours by modulating ECM expression and physical properties through the RhoA/ROCK signalling pathway. These valuable insights contribute to the development of combination therapies aimed at improving tumour targeting in cancer treatment.
Collapse
Affiliation(s)
- Xuedan Han
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Jialei Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yidong Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Eric Tse
- Sino Biopharmaceutical Group Limited, Beijing, 100026, China
| | - Qiyi Yu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yu Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi Ma
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
41
|
Wang J, Wang Y, Li J, Ying J, Mu Y, Zhang X, Zhou X, Sun L, Jiang H, Zhuo W, Shen Y, Zhou T, Liu X, Zhou Q. Neutrophil Extracellular Traps-Inhibiting and Fouling-Resistant Polysulfoxides Potently Prevent Postoperative Adhesion, Tumor Recurrence, and Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400894. [PMID: 38636448 DOI: 10.1002/adma.202400894] [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: 01/17/2024] [Revised: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Peritoneal metastasis (PM) is considered one of the most dreaded forms of cancer metastases for both patients and physicians. Aggressive cytoreductive surgery (CRS) is the primary treatment for peritoneal metastasis. Unfortunately, this intensive treatment frequently causes clinical complications, such as postoperative recurrence, metastasis, and adhesion formation. Emerging evidence suggests that neutrophil extracellular traps (NETs) released by inflammatory neutrophils contribute to these complications. Effective NET-targeting strategies thus show considerable potential in counteracting these complications but remain challenging. Here, one type of sulfoxide-containing homopolymer, PMeSEA, with potent fouling-resistant and NET-inhibiting capabilities, is synthesized and screened. Hydrating sulfoxide groups endow PMeSEA with superior nonfouling ability, significantly inhibiting protein/cell adhesion. Besides, the polysulfoxides can be selectively oxidized by ClO- which is required to stabilize the NETs rather than H2O2, and ClO- scavenging effectively inhibits NETs formation without disturbing redox homeostasis in tumor cells and quiescent neutrophils. As a result, PMeSEA potently prevents postoperative adhesions, significantly suppresses peritoneal metastasis, and shows synergetic antitumor activity with chemotherapeutic 5-Fluorouracil. Moreover, coupling CRS with PMeSEA potently inhibits CRS-induced tumor metastatic relapse and postoperative adhesions. Notably, PMeSEA exhibits low in vivo acute and subacute toxicities, implying significant potential for clinical postoperative adjuvant treatment.
Collapse
Affiliation(s)
- Jiafeng Wang
- Department of Pharmacology, and Department of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yechun Wang
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Junjun Li
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Jiajia Ying
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Yongli Mu
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Xuanhao Zhang
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Xuefei Zhou
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Leimin Sun
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Haiping Jiang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Wei Zhuo
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310020, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Tianhua Zhou
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310020, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Xiangrui Liu
- Department of Pharmacology, and Department of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310020, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Quan Zhou
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
42
|
He A, He L, Chen T, Li X, Cao C. Biomechanical Properties and Cellular Responses in Pulmonary Fibrosis. Bioengineering (Basel) 2024; 11:747. [PMID: 39199705 PMCID: PMC11351367 DOI: 10.3390/bioengineering11080747] [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: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Pulmonary fibrosis is a fatal lung disease affecting approximately 5 million people worldwide, with a 5-year survival rate of less than 50%. Currently, the only available treatments are palliative care and lung transplantation, as there is no curative drug for this condition. The disease involves the excessive synthesis of the extracellular matrix (ECM) due to alveolar epithelial cell damage, leading to scarring and stiffening of the lung tissue and ultimately causing respiratory failure. Although multiple factors contribute to the disease, the exact causes remain unclear. The mechanical properties of lung tissue, including elasticity, viscoelasticity, and surface tension, are not only affected by fibrosis but also contribute to its progression. This paper reviews the alteration in these mechanical properties as pulmonary fibrosis progresses and how cells in the lung, including alveolar epithelial cells, fibroblasts, and macrophages, respond to these changes, contributing to disease exacerbation. Furthermore, it highlights the importance of developing advanced in vitro models, based on hydrogels and 3D bioprinting, which can accurately replicate the mechanical and structural properties of fibrotic lungs and are conducive to studying the effects of mechanical stimuli on cellular responses. This review aims to summarize the current understanding of the interaction between the progression of pulmonary fibrosis and the alterations in mechanical properties, which could aid in the development of novel therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Andong He
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310028, China
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, 59 Liuting Road, Ningbo 315010, China
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Lizhe He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| | - Tianwei Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xuejin Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310028, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, 59 Liuting Road, Ningbo 315010, China
| |
Collapse
|
43
|
Ohnsorg ML, Mash KM, Khang A, Rao VV, Kirkpatrick BE, Bera K, Anseth KS. Nonlinear Elastic Bottlebrush Polymer Hydrogels Modulate Actomyosin Mediated Protrusion Formation in Mesenchymal Stromal Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403198. [PMID: 38655776 PMCID: PMC11239315 DOI: 10.1002/adma.202403198] [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: 03/01/2024] [Revised: 04/19/2024] [Indexed: 04/26/2024]
Abstract
The nonlinear elasticity of many tissue-specific extracellular matrices is difficult to recapitulate without the use of fibrous architectures, which couple strain-stiffening with stress relaxation. Herein, bottlebrush polymers are synthesized and crosslinked to form poly(ethylene glycol)-based hydrogels and used to study how strain-stiffening behavior affects human mesenchymal stromal cells (hMSCs). By tailoring the bottlebrush polymer length, the critical stress associated with the onset of network stiffening is systematically varied, and a unique protrusion-rich hMSC morphology emerges only at critical stresses within a biologically accessible stress regime. Local cell-matrix interactions are quantified using 3D traction force microscopy and small molecule inhibitors are used to identify cellular machinery that plays a critical role in hMSC mechanosensing of the engineered, strain-stiffening microenvironment. Collectively, this study demonstrates how covalently crosslinked bottlebrush polymer hydrogels can recapitulate strain-stiffening biomechanical cues at biologically relevant stresses and be used to probe how nonlinear elastic matrix properties regulate cellular processes.
Collapse
Affiliation(s)
- Monica L. Ohnsorg
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80308, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80308, USA
| | - Kayla M. Mash
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80308, USA
| | - Alex Khang
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80308, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80308, USA
| | - Varsha V. Rao
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80308, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80308, USA
| | - Bruce E. Kirkpatrick
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80308, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80308, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kaustav Bera
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80308, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80308, USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80308, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80308, USA
| |
Collapse
|
44
|
Vella MA, García DC, De Boeck M, Valdecantos PA, Roldán-Olarte M. Role of steroid hormones in the maintenance of focal adhesions in bovine oviductal epithelial cells. Domest Anim Endocrinol 2024; 88:106839. [PMID: 38433026 DOI: 10.1016/j.domaniend.2024.106839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/14/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
The oviduct, the organ of the female reproductive system where fertilization and early embryonic development occur, provides an optimal environment for the final maturation of oocytes, storage, and sperm capacitation and transport of gametes and embryos. During the estrous cycle, the oviduct is affected by ovarian sex hormones, resulting in changes aimed at maintaining an appropriate microenvironment. Normal cell migration is tightly regulated, its role being essential for the development and maintenance of organ and tissue functions as well as for regeneration following injury. Due to their involvement in focal contact formations, focal adhesion kinase (PTK2) and paxillin (PXN) are key proteins in the study of cell migration and adhesion. The objective of this work was to compare the expression of PTK2 and PXN in oviductal cells along the estrous cycle and to determine if their expression is regulated by the presence of 17-β estradiol (E2) and/or progesterone (P4). No transcripts of PTK2 or of PXN were detected in cells corresponding to the luteal phase. Additionally, hormonal stimulation experiments on bovine oviductal cell cultures (BOECs) were carried out, where P4 inhibited the expression of both genes. Migration assays demonstrated that P4 reduced BOECs migration capacity. P4 treatment also reduced cell adhesion, while E2 increased the number of adhered cells. In conclusion, the presence of E2 and P4 regulates the expression of genes involved in the formation of focal contacts and modifies the migration and adhesion of BOECs. Understanding the effect of steroid hormones on BOECs is critical to grasp the impact of steroid control on oviductal function and its contribution to establishing successful pregnancies.
Collapse
Affiliation(s)
- Milda Alejandra Vella
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Biología 'Dr. Francisco D. Barbieri', Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Chacabuco 461, T4000ILI San Miguel de Tucumán, Tucumán, Argentina
| | - Daniela Celeste García
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Biología 'Dr. Francisco D. Barbieri', Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Chacabuco 461, T4000ILI San Miguel de Tucumán, Tucumán, Argentina; Instituto de Ciencias Químicas (ICQ), Facultad de Agronomía y Agroindustrias (FAyA), Universidad Nacional de Santiago del Estero (UNSE)-CONICET. RN 9 Km 1125. Villa El Zanjón. CP4206 Santiago del Estero, Argentina
| | - Maximiliano De Boeck
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Biología 'Dr. Francisco D. Barbieri', Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Chacabuco 461, T4000ILI San Miguel de Tucumán, Tucumán, Argentina
| | - Pablo Alberto Valdecantos
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Biología 'Dr. Francisco D. Barbieri', Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Chacabuco 461, T4000ILI San Miguel de Tucumán, Tucumán, Argentina
| | - Mariela Roldán-Olarte
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Biología 'Dr. Francisco D. Barbieri', Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Chacabuco 461, T4000ILI San Miguel de Tucumán, Tucumán, Argentina.
| |
Collapse
|
45
|
Sun P, Li Y, Yu W, Chen J, Wan P, Wang Z, Zhang M, Wang C, Fu S, Mang G, Choi S, Du Z, Tang C, Li S, Shi G, Tian J, Dai J, Leng X. Low-intensity pulsed ultrasound improves myocardial ischaemia‒reperfusion injury via migrasome-mediated mitocytosis. Clin Transl Med 2024; 14:e1749. [PMID: 38951127 PMCID: PMC11216834 DOI: 10.1002/ctm2.1749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
During myocardial ischaemia‒reperfusion injury (MIRI), the accumulation of damaged mitochondria could pose serious threats to the heart. The migrasomes, newly discovered mitocytosis-mediating organelles, selectively remove damaged mitochondria to provide mitochondrial quality control. Here, we utilised low-intensity pulsed ultrasound (LIPUS) on MIRI mice model and demonstrated that LIPUS reduced the infarcted area and improved cardiac dysfunction. Additionally, we found that LIPUS alleviated MIRI-induced mitochondrial dysfunction. We provided new evidence that LIPUS mechanical stimulation facilitated damaged mitochondrial excretion via migrasome-dependent mitocytosis. Inhibition the formation of migrasomes abolished the protective effect of LIPUS on MIRI. Mechanistically, LIPUS induced the formation of migrasomes by evoking the RhoA/Myosin II/F-actin pathway. Meanwhile, F-actin activated YAP nuclear translocation to transcriptionally activate the mitochondrial motor protein KIF5B and Drp1, which are indispensable for LIPUS-induced mitocytosis. These results revealed that LIPUS activates mitocytosis, a migrasome-dependent mitochondrial quality control mechanism, to protect against MIRI, underlining LIPUS as a safe and potentially non-invasive treatment for MIRI.
Collapse
Affiliation(s)
- Ping Sun
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
| | - Yifei Li
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
| | - Weidong Yu
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
| | - Jianfeng Chen
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
- Laboratory of Animal CenterThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Pingping Wan
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Zhuo Wang
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
| | - Maomao Zhang
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Chao Wang
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
| | - Shuai Fu
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
| | - Ge Mang
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Stephen Choi
- SXULTRASONIC Ltd. Kerry Rehabilitation Medicine Research InstituteShenzhenChina
| | - Zhuo Du
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Caiying Tang
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Song Li
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Guoxia Shi
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Jiawei Tian
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
| | - Jiannan Dai
- The Key Laboratory of Myocardial IschemiaHarbin Medical University, Ministry of EducationHarbinChina
- Department of CardiologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Xiaoping Leng
- Department of UltrasoundThe Second Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang ProvinceHarbinChina
| |
Collapse
|
46
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
47
|
Miller EM, Chan TCD, Montes-Matamoros C, Sharif O, Pujo-Menjouet L, Lindstrom MR. Oscillations in Neuronal Activity: A Neuron-Centered Spatiotemporal Model of the Unfolded Protein Response in Prion Diseases. Bull Math Biol 2024; 86:82. [PMID: 38837083 DOI: 10.1007/s11538-024-01307-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
Many neurodegenerative diseases (NDs) are characterized by the slow spatial spread of toxic protein species in the brain. The toxic proteins can induce neuronal stress, triggering the Unfolded Protein Response (UPR), which slows or stops protein translation and can indirectly reduce the toxic load. However, the UPR may also trigger processes leading to apoptotic cell death and the UPR is implicated in the progression of several NDs. In this paper, we develop a novel mathematical model to describe the spatiotemporal dynamics of the UPR mechanism for prion diseases. Our model is centered around a single neuron, with representative proteins P (healthy) and S (toxic) interacting with heterodimer dynamics (S interacts with P to form two S's). The model takes the form of a coupled system of nonlinear reaction-diffusion equations with a delayed, nonlinear flux for P (delay from the UPR). Through the delay, we find parameter regimes that exhibit oscillations in the P- and S-protein levels. We find that oscillations are more pronounced when the S-clearance rate and S-diffusivity are small in comparison to the P-clearance rate and P-diffusivity, respectively. The oscillations become more pronounced as delays in initiating the UPR increase. We also consider quasi-realistic clinical parameters to understand how possible drug therapies can alter the course of a prion disease. We find that decreasing the production of P, decreasing the recruitment rate, increasing the diffusivity of S, increasing the UPR S-threshold, and increasing the S clearance rate appear to be the most powerful modifications to reduce the mean UPR intensity and potentially moderate the disease progression.
Collapse
Affiliation(s)
- Elliot M Miller
- College of Arts and Sciences, Culverhouse College of Business, The University of Alabama, Tuscaloosa, AL, USA
| | - Tat Chung D Chan
- Department of Mathematics, University of California Berkeley, Berkeley, CA, USA
| | - Carlos Montes-Matamoros
- School of Mathematical and Statistical Sciences, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Omar Sharif
- School of Mathematical and Statistical Sciences, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Laurent Pujo-Menjouet
- Universite Claude Bernard Lyon 1, CNRS, Ecole Centrale de Lyon, INSA Lyon, Université Jean Monnet, ICJ UMR5208, Inria, 69622, Villeurbanne, France
| | - Michael R Lindstrom
- School of Mathematical and Statistical Sciences, The University of Texas Rio Grande Valley, Edinburg, TX, USA.
| |
Collapse
|
48
|
Chen H, Wang S, Cao Y, Lei H. Molecular Force Sensors for Biological Application. Int J Mol Sci 2024; 25:6198. [PMID: 38892386 PMCID: PMC11173168 DOI: 10.3390/ijms25116198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The mechanical forces exerted by cells on their surrounding microenvironment are known as cellular traction forces. These forces play crucial roles in various biological processes, such as tissue development, wound healing and cell functions. However, it is hard for traditional techniques to measure cellular traction forces accurately because their magnitude (from pN to nN) and the length scales over which they occur (from nm to μm) are extremely small. In order to fully understand mechanotransduction, highly sensitive tools for measuring cellular forces are needed. Current powerful techniques for measuring traction forces include traction force microscopy (TFM) and fluorescent molecular force sensors (FMFS). In this review, we elucidate the force imaging principles of TFM and FMFS. Then we highlight the application of FMFS in a variety of biological processes and offer our perspectives and insights into the potential applications of FMFS.
Collapse
Affiliation(s)
- Huiyan Chen
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Shouhan Wang
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Hai Lei
- School of Physics, Zhejiang University, Hangzhou 310027, China
- Institute for Advanced Study in Physics, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
49
|
Lee ST, Kuboki T, Kidoaki S, Aida Y, Arima Y, Tamada K. A plasmonic metasurface reveals differential motility of breast cancer cell lines at initial phase of adhesion. Colloids Surf B Biointerfaces 2024; 238:113876. [PMID: 38555764 DOI: 10.1016/j.colsurfb.2024.113876] [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: 01/31/2024] [Revised: 03/17/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
A plasmonic metasurface composed of a self-assembled monolayer of gold nanoparticles allows for fluorescence imaging with high spatial resolution, owing to the collective excitation of localized surface plasmon resonance. Taking advantage of fluorescence imaging confined to the nano-interface, we examined actin organization in breast cancer cell lines with different metastatic potentials during cell adhesion. Live-cell fluorescence imaging confined within tens of nanometers from the substrate shows a high actin density spanning < 1 μm from the cell edge. Live-cell imaging revealed that the breast cancer cell lines exhibited different actin patterns during the initial phase of cell adhesion (∼ 1 h). Non-tumorous MCF10A cells exhibited symmetric actin localization at the cell edge, whereas highly metastatic MDA-MB-231 cells showed asymmetric actin localization, demonstrating rapid polarization of MDA-MB-231 cells upon adhesion. The rapid actin organization observed by our plasmonic metasurface-based fluorescence imaging provides information on how quickly cancer cells sense the underlying substrate.
Collapse
Affiliation(s)
- Shi Ting Lee
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Thasaneeya Kuboki
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Satoru Kidoaki
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yukiko Aida
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yusuke Arima
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| | - Kaoru Tamada
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
50
|
Yang Z, Peng Y, Wang Y, Yang P, Huang Z, Quan T, Xu X, Sun P, Sun Y, Lv J, Wei D, Zhou GQ. KLF5 regulates actin remodeling to enhance the metastasis of nasopharyngeal carcinoma. Oncogene 2024; 43:1779-1795. [PMID: 38649438 DOI: 10.1038/s41388-024-03033-0] [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: 09/26/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Transcription factors (TFs) engage in various cellular essential processes including differentiation, growth and migration. However, the master TF involved in distant metastasis of nasopharyngeal carcinoma (NPC) remains largely unclear. Here we show that KLF5 regulates actin remodeling to enhance NPC metastasis. We analyzed the msVIPER algorithm-generated transcriptional regulatory networks and identified KLF5 as a master TF of metastatic NPC linked to poor clinical outcomes. KLF5 regulates actin remodeling and lamellipodia formation to promote the metastasis of NPC cells in vitro and in vivo. Mechanistically, KLF5 preferentially occupies distal enhancer regions of ACTN4 to activate its transcription, whereby decoding the informative DNA sequences. ACTN4, extensively localized within actin cytoskeleton, facilitates dense and branched actin networks and lamellipodia formation at the cell leading edge, empowering cells to migrate faster. Collectively, our findings reveal that KLF5 controls robust transcription program of ACTN4 to modulate actin remodeling and augment cell motility which enhances NPC metastasis, and provide new potential biomarkers and therapeutic interventions for NPC.
Collapse
Affiliation(s)
- Zhenyu Yang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Yanfu Peng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Yaqin Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Panyang Yang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Zhuohui Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Tingqiu Quan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Xudong Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Peng Sun
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Ying Sun
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Jiawei Lv
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| | - Denghui Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| | - Guan-Qun Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China.
| |
Collapse
|