1
|
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
|
2
|
Morishita H, Kawai K, Egami Y, Honda K, Araki N. Live-cell imaging and CLEM reveal the existence of ACTN4-dependent ruffle-edge lamellipodia acting as a novel mode of cell migration. Exp Cell Res 2024; 442:114232. [PMID: 39222868 DOI: 10.1016/j.yexcr.2024.114232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
α-Actinin-4 (ACTN4) expression levels are correlated with the invasive and metastatic potential of cancer cells; however, the underlying mechanism remains unclear. Here, we identified ACTN4-localized ruffle-edge lamellipodia using live-cell imaging and correlative light and electron microscopy (CLEM). BSC-1 cells expressing EGFP-ACTN4 showed that ACTN4 was most abundant in the leading edges of lamellipodia, although it was also present in stress fibers and focal adhesions. ACTN4 localization in lamellipodia was markedly diminished by phosphoinositide 3-kinase inhibition, whereas its localization in stress fibers and focal adhesions remained. Furthermore, overexpression of ACTN4, but not ACTN1, promoted lamellipodial formation. Live-cell analysis demonstrated that ACTN4-enriched lamellipodia are highly dynamic and associated with cell migration. CLEM revealed that ACTN4-enriched lamellipodia exhibit a characteristic morphology of multilayered ruffle-edges that differs from canonical flat lamellipodia. Similar ruffle-edge lamellipodia were observed in A549 and MDA-MB-231 invasive cancer cells. ACTN4 knockdown suppressed the formation of ruffle-edge lamellipodia and cell migration during wound healing in A549 monolayer cultures. Additionally, membrane-type 1 matrix metalloproteinase was observed in the membrane ruffles, suggesting that ruffle-edge lamellipodia have the ability to degrade the extracellular matrix and may contribute to active cell migration/invasion in certain cancer cell types.
Collapse
Affiliation(s)
- Haruka Morishita
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Katsuhisa Kawai
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Youhei Egami
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Kazufumi Honda
- Department of Bioregulation, Graduate of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, 113-8602, Tokyo, Japan
| | - Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan.
| |
Collapse
|
3
|
Chang SL, Tsai YJ, Shieh JM, Wu WB. The novel thromboxane prostanoid receptor mediates CTGF production to drive human nasal fibroblast self-migration through NF-κB and PKCδ-CREB signaling pathways. J Cell Physiol 2024; 239:e31390. [PMID: 39104040 DOI: 10.1002/jcp.31390] [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: 03/11/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
Chronic rhinosinusitis without nasal polyp (CRSsNP) is characterized by tissue repair/remodeling and the subepithelial stroma region in whose nasal mucosa has been reported by us to have thromboxane A2 (TXA2) prostanoid (TP) receptor and overexpress connective tissue growth factor (CTGF). Therefore, this study aimed to investigate the relationship between TP receptor activation and CTGF production/function in human CRSsNP nasal mucosa stromal fibroblasts. We found that TP agonists including U46619 and IBOP ([1S-[1α,2α(Z),3β(1E,3 S*),4α]]-7-[3-[3-hydroxy-4-(4-iodophenoxy)-1-butenyl]-7-oxabicyclo[2.2.1]hept-2-yl]-5-heptenoic acid) could promote CTGF protein/messenger RNA expression and secretion. The pharmacological intervention and TP activation assay with U46619 identified the possible participation of PKCμ, PKCδ, nuclear factor-κB (NF-κB), and cyclic AMP response element-binding protein (CREB) phosphorylation/activation in the CTGF induction. Moreover, a phorbol ester-phorbol-12-myristate 13-acetate (PMA) exhibited a similar cellular signaling and CTGF production profile to that elicited by TP activation. However, further small interfering RNA interference analysis revealed that only NF-κB and PKCδ-CREB pathways were necessarily required for TP-mediated CTGF production, which could not be completely supported by those findings from PMA. Finally, in a functional assay, although CTGF did not affect fibroblast proliferation, TP-mediated CTGF could drive novel self-migration in fibroblasts both in the scratch/wound healing and transwell apparatus assays. Meanwhile, the overall staining for stress fibers and formation of the lamellipodia and filopodia-like structures was concomitantly increased in the treated migrating cells. Collectively, we provided here that novel TP mediates CTGF production and self-migration in human nasal fibroblasts through NF-κB and PKCδ-CREB signaling pathways. More importantly, we also demonstrated that thromboxane, TP receptor, CTGF, and stromal fibroblasts may act in concert in the tissue remodeling/repair process during CRSsNP development and progression.
Collapse
Affiliation(s)
- Shih-Lun Chang
- Department of Otorhinolaryngology, Chi Mei Medical Center, Yongkang District, Tainan, Taiwan
- Department of Pet Care and Grooming, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Yih-Jeng Tsai
- Department of Otolaryngology Head and Neck Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Jiunn-Min Shieh
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Wen-Bin Wu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
4
|
Iftikhar A, Shepherd S, Jones S, Ellis I. Effect of Mifepristone on Migration and Proliferation of Oral Cancer Cells. Int J Mol Sci 2024; 25:8777. [PMID: 39201464 PMCID: PMC11354386 DOI: 10.3390/ijms25168777] [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: 06/07/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Glucocorticoid receptor (GR) overexpression has been linked to increased tumour aggressiveness and treatment resistance. GR antagonists have been shown to enhance treatment effectiveness. Emerging research has investigated mifepristone, a GR antagonist, as an anticancer agent with limited research in the context of oral cancer. This study investigated the effect of mifepristone at micromolar (µM) concentrations of 1, 5, 10 and 20 on the proliferation and migration of oral cancer cells, at 24 and 48 h. Scratch and scatter assays were utilised to assess cell migration, MTT assays were used to measure cell proliferation, Western blotting was used to investigate the expression of GR and the activation of underlying Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) signalling pathways, and immunofluorescence (IF) was used to determine the localisation of proteins in HaCaT (immortalised human skin keratinocytes), TYS (oral adeno squamous cell carcinoma), and SAS-H1 cells (squamous cell carcinoma of human tongue). Mifepristone resulted in a dose-dependent reduction in the proliferation of HaCaT, TYS, and SAS-H1 cells. Mifepristone at a concentration of 20 µM effectively reduced collective migration and scattering of oral cancer cells, consistent with the suppression of the PI3K-Akt and MAPK signalling pathways, and reduced expression of N-Cadherin. An elongated cell morphology was, however, observed, which may be linked to the localisation pattern of E-Cadherin in response to mifepristone. Overall, this study found that a high concentration of mifepristone was effective in the suppression of migration and proliferation of oral cancer cells via the inhibition of PI3K-Akt and MAPK signalling pathways. Further investigation is needed to define its impact on epithelial-mesenchymal transition (EMT) markers.
Collapse
Affiliation(s)
| | | | | | - Ian Ellis
- School of Dentistry, University of Dundee, Dundee DD1 4HR, UK; (A.I.); (S.S.); (S.J.)
| |
Collapse
|
5
|
Fresquez AM, Hogan JO, Rivera P, Patterson KM, Singer K, Reynolds JM, White C. STIM1-dependent store-operated calcium entry mediates sex differences in macrophage chemotaxis and monocyte recruitment. J Biol Chem 2024; 300:107422. [PMID: 38815866 PMCID: PMC11231831 DOI: 10.1016/j.jbc.2024.107422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 05/09/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
Infiltration of monocyte-derived cells to sites of infection and injury is greater in males than in females, due in part, to increased chemotaxis, the process of directed cell movement toward a chemical signal. The mechanisms governing sexual dimorphism in chemotaxis are not known. We hypothesized a role for the store-operated calcium entry (SOCE) pathway in regulating chemotaxis by modulating leading and trailing edge membrane dynamics. We measured the chemotactic response of bone marrow-derived macrophages migrating toward complement component 5a (C5a). Chemotactic ability was dependent on sex and inflammatory phenotype (M0, M1, and M2), and correlated with SOCE. Notably, females exhibited a significantly lower magnitude of SOCE than males. When we knocked out the SOCE gene, stromal interaction molecule 1 (STIM1), it eliminated SOCE and equalized chemotaxis across both sexes. Analysis of membrane dynamics at the leading and trailing edges showed that STIM1 influences chemotaxis by facilitating retraction of the trailing edge. Using BTP2 to pharmacologically inhibit SOCE mirrored the effects of STIM1 knockout, demonstrating a central role of STIM/Orai-mediated calcium signaling. Importantly, by monitoring the recruitment of adoptively transferred monocytes in an in vivo model of peritonitis, we show that increased infiltration of male monocytes during infection is dependent on STIM1. These data support a model in which STIM1-dependent SOCE is necessary and sufficient for mediating the sex difference in monocyte recruitment and macrophage chemotactic ability by regulating trailing edge dynamics.
Collapse
Affiliation(s)
- Adriana M Fresquez
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - James O Hogan
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Patricia Rivera
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Kristen M Patterson
- Microbiology and Immunology, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph M Reynolds
- Microbiology and Immunology, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Carl White
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA.
| |
Collapse
|
6
|
Quinsgaard EMB, Korsnes MS, Korsnes R, Moestue SA. Single-cell tracking as a tool for studying EMT-phenotypes. Exp Cell Res 2024; 437:113993. [PMID: 38485079 DOI: 10.1016/j.yexcr.2024.113993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/24/2024]
Abstract
This article demonstrates that label-free single-cell video tracking is a useful approach for in vitro studies of Epithelial-Mesenchymal Transition (EMT). EMT is a highly heterogeneous process, involved in wound healing, embryogenesis and cancer. The process promotes metastasis, and increased understanding can aid development of novel therapeutic strategies. The role of EMT-associated biomarkers depends on biological context, making it challenging to compare and interpret data from different studies. We demonstrate single-cell video tracking for comprehensive phenotype analysis. In this study we performed single-cell video tracking on 72-h long recordings. We quantified several behaviours at a single-cell level during induced EMT in MDA-MB-468 cells. This revealed notable variations in migration speed, with different dose-response patterns and varying distributions of speed. By registering cell morphologies during the recording, we determined preferred paths of morphological transitions. We also found a clear association between migration speed and cell morphology. We found elevated rates of cell death, diminished proliferation, and an increase in mitotic failures followed by re-fusion of sister-cells. The method allows tracking of phenotypes in cell lineages, which can be particularly useful in epigenetic studies. Sister-cells were found to have significant similarities in their speeds and morphologies, illustrating the heritability of these traits.
Collapse
Affiliation(s)
- Ellen Marie Botne Quinsgaard
- Norwegian University of Science and Technology (NTNU), Department of Clinical and Molecular Medicine, NO-7491 Trondheim, Norway.
| | - Mónica Suárez Korsnes
- Norwegian University of Science and Technology (NTNU), Department of Clinical and Molecular Medicine, NO-7491 Trondheim, Norway; Korsnes Biocomputing (KoBio), Trondheim, Norway
| | | | - Siver Andreas Moestue
- Norwegian University of Science and Technology (NTNU), Department of Clinical and Molecular Medicine, NO-7491 Trondheim, Norway; Department of Pharmacy, Nord University, Bodø, Norway
| |
Collapse
|
7
|
Arce FT, Younger S, Gaber AA, Mascarenhas JB, Rodriguez M, Dudek SM, Garcia JGN. Lamellipodia dynamics and microrheology in endothelial cell paracellular gap closure. Biophys J 2023; 122:4730-4747. [PMID: 37978804 PMCID: PMC10754712 DOI: 10.1016/j.bpj.2023.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/06/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023] Open
Abstract
Vascular endothelial cells (ECs) form a semipermeable barrier separating vascular contents from the interstitium, thereby regulating the movement of water and molecular solutes across small intercellular gaps, which are continuously forming and closing. Under inflammatory conditions, however, larger EC gaps form resulting in increased vascular leakiness to circulating fluid, proteins, and cells, which results in organ edema and dysfunction responsible for key pathophysiologic findings in numerous inflammatory disorders. In this study, we extend our earlier work examining the biophysical properties of EC gap formation and now address the role of lamellipodia, thin sheet-like membrane projections from the leading edge, in modulating EC spatial-specific contractile properties and gap closure. Micropillars, fabricated by soft lithography, were utilized to form reproducible paracellular gaps in human lung ECs. Using time-lapse imaging via optical microscopy, rates of EC gap closure and motility were measured with and without EC stimulation with the barrier-enhancing sphingolipid, sphingosine-1-phosphate. Peripheral ruffle formation was ubiquitous during gap closure. Kymographs were generated to quantitatively compare the lamellipodia dynamics of sphingosine-1-phosphate-stimulated and -unstimulated ECs. Utilizing atomic force microscopy, we characterized the viscoelastic behavior of EC lamellipodia. Our results indicate decreased stiffness and increased liquid-like behavior of expanding lamellipodia compared with regions away from the cellular edge (lamella and cell body) during EC gap closure, results in sync with the rapid kinetics of protrusion/retraction motion. We hypothesize this dissipative EC behavior during gap closure is linked to actomyosin cytoskeletal rearrangement and decreased cross-linking during lamellipodia expansion. In summary, these studies of the kinetic and mechanical properties of EC lamellipodia and ruffles at gap boundaries yield insights into the mechanisms of vascular barrier restoration and potentially a model system for examining the druggability of lamellipodial protein targets to enhance vascular barrier integrity.
Collapse
Affiliation(s)
- Fernando Teran Arce
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida.
| | - Scott Younger
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Amir A Gaber
- Department of Medicine, University of Arizona, Tucson, Arizona
| | | | - Marisela Rodriguez
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida; Department of Medicine, University of Arizona, Tucson, Arizona
| | - Steven M Dudek
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois
| | - Joe G N Garcia
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, Florida.
| |
Collapse
|
8
|
Leineweber WD, Fraley SI. Adhesion tunes speed and persistence by coordinating protrusions and extracellular matrix remodeling. Dev Cell 2023; 58:1414-1428.e4. [PMID: 37321214 PMCID: PMC10527808 DOI: 10.1016/j.devcel.2023.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 03/14/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023]
Abstract
Cell migration through 3D environments is essential to development, disease, and regeneration processes. Conceptual models of migration have been developed primarily on the basis of 2D cell behaviors, but a general understanding of 3D cell migration is still lacking due to the added complexity of the extracellular matrix. Here, using a multiplexed biophysical imaging approach for single-cell analysis of human cell lines, we show how the subprocesses of adhesion, contractility, actin cytoskeletal dynamics, and matrix remodeling integrate to produce heterogeneous migration behaviors. This single-cell analysis identifies three modes of cell speed and persistence coupling, driven by distinct modes of coordination between matrix remodeling and protrusive activity. The framework that emerges establishes a predictive model linking cell trajectories to distinct subprocess coordination states.
Collapse
Affiliation(s)
- William D Leineweber
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephanie I Fraley
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
9
|
Zhou FY, Weems A, Gihana GM, Chen B, Chang BJ, Driscoll M, Danuser G. Surface-guided computing to analyze subcellular morphology and membrane-associated signals in 3D. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536640. [PMID: 37131779 PMCID: PMC10153113 DOI: 10.1101/2023.04.12.536640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Signal transduction and cell function are governed by the spatiotemporal organization of membrane-associated molecules. Despite significant advances in visualizing molecular distributions by 3D light microscopy, cell biologists still have limited quantitative understanding of the processes implicated in the regulation of molecular signals at the whole cell scale. In particular, complex and transient cell surface morphologies challenge the complete sampling of cell geometry, membrane-associated molecular concentration and activity and the computing of meaningful parameters such as the cofluctuation between morphology and signals. Here, we introduce u-Unwrap3D, a framework to remap arbitrarily complex 3D cell surfaces and membrane-associated signals into equivalent lower dimensional representations. The mappings are bidirectional, allowing the application of image processing operations in the data representation best suited for the task and to subsequently present the results in any of the other representations, including the original 3D cell surface. Leveraging this surface-guided computing paradigm, we track segmented surface motifs in 2D to quantify the recruitment of Septin polymers by blebbing events; we quantify actin enrichment in peripheral ruffles; and we measure the speed of ruffle movement along topographically complex cell surfaces. Thus, u-Unwrap3D provides access to spatiotemporal analyses of cell biological parameters on unconstrained 3D surface geometries and signals.
Collapse
Affiliation(s)
- Felix Y. Zhou
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Weems
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabriel M. Gihana
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bingying Chen
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bo-Jui Chang
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meghan Driscoll
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Current address: Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. & Ida Green Center for System Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
10
|
Powers RM, Hevner RF, Halpain S. The Neuron Navigators: Structure, function, and evolutionary history. Front Mol Neurosci 2023; 15:1099554. [PMID: 36710926 PMCID: PMC9877351 DOI: 10.3389/fnmol.2022.1099554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Neuron navigators (Navigators) are cytoskeletal-associated proteins important for neuron migration, neurite growth, and axon guidance, but they also function more widely in other tissues. Recent studies have revealed novel cellular functions of Navigators such as macropinocytosis, and have implicated Navigators in human disorders of axon growth. Navigators are present in most or all bilaterian animals: vertebrates have three Navigators (NAV1-3), Drosophila has one (Sickie), and Caenorhabditis elegans has one (Unc-53). Structurally, Navigators have conserved N- and C-terminal regions each containing specific domains. The N-terminal region contains a calponin homology (CH) domain and one or more SxIP motifs, thought to interact with the actin cytoskeleton and mediate localization to microtubule plus-end binding proteins, respectively. The C-terminal region contains two coiled-coil domains, followed by a AAA+ family nucleoside triphosphatase domain of unknown activity. The Navigators appear to have evolved by fusion of N- and C-terminal region homologs present in simpler organisms. Overall, Navigators participate in the cytoskeletal response to extracellular cues via microtubules and actin filaments, in conjunction with membrane trafficking. We propose that uptake of fluid-phase cues and nutrients and/or downregulation of cell surface receptors could represent general mechanisms that explain Navigator functions. Future studies developing new models, such as conditional knockout mice or human cerebral organoids may reveal new insights into Navigator function. Importantly, further biochemical studies are needed to define the activities of the Navigator AAA+ domain, and to study potential interactions among different Navigators and their binding partners.
Collapse
Affiliation(s)
- Regina M. Powers
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Robert F. Hevner
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,Department of Pathology, UC San Diego School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Shelley Halpain
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, United States,Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States,*Correspondence: Shelley Halpain, ✉
| |
Collapse
|
11
|
Angeloni L, Popa B, Nouri-Goushki M, Minneboo M, Zadpoor AA, Ghatkesar MK, Fratila-Apachitei LE. Fluidic Force Microscopy and Atomic Force Microscopy Unveil New Insights into the Interactions of Preosteoblasts with 3D-Printed Submicron Patterns. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204662. [PMID: 36373704 DOI: 10.1002/smll.202204662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Physical patterns represent potential surface cues for promoting osteogenic differentiation of stem cells and improving osseointegration of orthopedic implants. Understanding the early cell-surface interactions and their effects on late cellular functions is essential for a rational design of such topographies, yet still elusive. In this work, fluidic force microscopy (FluidFM) and atomic force microscopy (AFM) combined with optical and electron microscopy are used to quantitatively investigate the interaction of preosteoblasts with 3D-printed patterns after 4 and 24 h of culture. The patterns consist of pillars with the same diameter (200 nm) and interspace (700 nm) but distinct heights (500 and 1000 nm) and osteogenic properties. FluidFM reveals a higher cell adhesion strength after 24 h of culture on the taller pillars (32 ± 7 kPa versus 21.5 ± 12.5 kPa). This is associated with attachment of cells partly on the sidewalls of these pillars, thus requiring larger normal forces for detachment. Furthermore, the higher resistance to shear forces observed for these cells indicates an enhanced anchorage and can be related to the persistence and stability of lamellipodia. The study explains the differential cell adhesion behavior induced by different pillar heights, enabling advancements in the rational design of osteogenic patterns.
Collapse
Affiliation(s)
- Livia Angeloni
- Department of Precision and Microsystems Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Bogdan Popa
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Mahdiyeh Nouri-Goushki
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Michelle Minneboo
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Murali K Ghatkesar
- Department of Precision and Microsystems Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Lidy E Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| |
Collapse
|
12
|
Ritch SJ, Noman ASM, Goyeneche AA, Telleria CM. The metastatic capacity of high-grade serous ovarian cancer cells changes along disease progression: inhibition by mifepristone. Cancer Cell Int 2022; 22:397. [PMID: 36494669 PMCID: PMC9733158 DOI: 10.1186/s12935-022-02822-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Simplistic two-dimensional (2D) in vitro assays have long been the standard for studying the metastatic abilities of cancer cells. However, tri-dimensional (3D) organotypic models provide a more complex environment, closer to that seen in patients, and thereby provide a more accurate representation of their true capabilities. Our laboratory has previously shown that the antiprogestin and antiglucocorticoid mifepristone can reduce the growth, adhesion, migration, and invasion of various aggressive cancer cells assessed using 2D assays. In this study, we characterize the metastatic capabilities of high-grade serous ovarian cancer cells generated along disease progression, in both 2D and 3D assays, and the ability of cytostatic doses of mifepristone to inhibit them. METHODS High-grade serous ovarian cancer cells collected from two separate patients at different stages of their disease were used throughout the study. The 2D wound healing and Boyden chamber assays were used to study migration, while a layer of extracellular matrix was added to the Boyden chamber to study invasion. A 3D organotypic model, composed of fibroblasts embedded in collagen I and topped with a monolayer of mesothelial cells was used to further study cancer cell adhesion and mesothelial displacement. All assays were studied in cells, which were originally harvested from two patients at different stages of disease progression, in the absence or presence of cytostatic doses of mifepristone. RESULTS 2D in vitro assays demonstrated that the migration and invasive rates of the cells isolated from both patients decreased along disease progression. Conversely, in both patients, cells representing late-stage disease demonstrated a higher adhesion capacity to the 3D organotypic model than those representing an early-stage disease. This adhesive behavior is associated with the in vivo tumor capacity of the cells. Regardless of these differences in adhesive, migratory, and invasive behavior among the experimental protocols used, cytostatic doses of mifepristone were able to inhibit the adhesion, migration, and invasion rates of all cells studied, regardless of their basal capabilities over simplistic or organotypic metastatic in vitro model systems. Finally, we demonstrate that when cells acquire the capacity to grow spontaneously as spheroids, they do attach to a 3D organotypic model system when pre-incubated with conditioned media. Of relevance, mifepristone was able to cause dissociation of these multicellular structures. CONCLUSION Differences in cellular behaviours were observed between 2 and 3D assays when studying the metastatic capabilities of high-grade serous ovarian cancer cells representing disease progression. Mifepristone inhibited these metastatic capabilities in all assays studied.
Collapse
Affiliation(s)
- Sabrina J. Ritch
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada
| | - Abu Shadat M. Noman
- grid.413089.70000 0000 9744 3393Department of Biochemistry and Molecular Biology, Chittagong University, Chittagong, Bangladesh
| | - Alicia A. Goyeneche
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada ,grid.63984.300000 0000 9064 4811Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC Canada
| | - Carlos M. Telleria
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada ,grid.63984.300000 0000 9064 4811Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC Canada
| |
Collapse
|
13
|
Liu L, Sui R, Li L, Zhang L, Zeng D, Ni X, Sun J. Light Activates Cdc42-Mediated Needle-Shaped Filopodia Formation via the Integration of Small GTPases. Cell Mol Bioeng 2022; 15:599-609. [PMID: 36531863 PMCID: PMC9751244 DOI: 10.1007/s12195-022-00743-x] [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: 03/14/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction Cdc42 has been linked to multiple human cancers and is implicated in the migration of cancer cells. Cdc42 could be activated via biochemical and biophysical factors in tumor microenvironment, the precise control of Cdc42 was essential to determine its role to cell behaviors. Needle-shaped protrusions (filopodia) could sense the extracellular biochemical cues and pave the path for cell movement, which was a key structure involved in the regulation of cancer cell motility. Methods We used the photoactivatable Cdc42 to elucidate the breast cancer cell protrusions, the mutation of Cdc42 was to confirm the optogenetic results. We also inhibit the Cdc42, Rac or Rho respectively by the corresponding inhibitors. Results We identified that the activation of Cdc42 by light could greatly enhance the formation of filopodia, which was positive for the contribution of cell movement. The expression of Cdc42 active form Cdc42-Q61L in cells resulted in the longer and more filopodia while the Cdc42 inactive form Cdc42-T17N were with the shorter and less filopodia. Moreover, the inhibition of Cdc42, Rac or Rho all significantly reduced the filopodia numbers and length in the co-expression of Cdc42-Q61L, which showed that the integration of small GTPases was necessary in the formation of filopodia. Furthermore, photoactivation of Cdc42 failed to enhance the filopodia formation with the inhibition of Rac or Rho. However, with the inhibition of Cdc42, the photoactivation of Cdc42 could partially recover back the filopodia formations, which indicated that the integration of small GTPases was key for the filopodia formations. Conclusions Our work highlights that light activates Cdc42 is sufficient to promote filopodia formation without the destructive structures of small GTPases, it not only points out the novel technique to determine cell structure formations but also provides the experimental basis for the efficient small GTPases-based anti-cancer strategies.
Collapse
Affiliation(s)
- Lingling Liu
- School of Medical Laboratory Science, Chengdu Medical College, Chengdu, 610500 Sichuan China
| | - Ran Sui
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan China
| | - Lianxin Li
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan China
| | - Lin Zhang
- School of Medical Laboratory Science, Chengdu Medical College, Chengdu, 610500 Sichuan China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan China
| | - Jinghui Sun
- School of Medical Laboratory Science, Chengdu Medical College, Chengdu, 610500 Sichuan China
| |
Collapse
|
14
|
Brosch PK, Korsa T, Taban D, Eiring P, Kreisz P, Hildebrand S, Neubauer J, Zimmermann H, Sauer M, Shirakashi R, Djuzenova CS, Sisario D, Sukhorukov VL. Glucose and Inositol Transporters, SLC5A1 and SLC5A3, in Glioblastoma Cell Migration. Cancers (Basel) 2022; 14:5794. [PMID: 36497276 PMCID: PMC9738886 DOI: 10.3390/cancers14235794] [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: 10/06/2022] [Revised: 11/04/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
(1) Background: The recurrence of glioblastoma multiforme (GBM) is mainly due to invasion of the surrounding brain tissue, where organic solutes, including glucose and inositol, are abundant. Invasive cell migration has been linked to the aberrant expression of transmembrane solute-linked carriers (SLC). Here, we explore the role of glucose (SLC5A1) and inositol transporters (SLC5A3) in GBM cell migration. (2) Methods: Using immunofluorescence microscopy, we visualized the subcellular localization of SLC5A1 and SLC5A3 in two highly motile human GBM cell lines. We also employed wound-healing assays to examine the effect of SLC inhibition on GBM cell migration and examined the chemotactic potential of inositol. (3) Results: While GBM cell migration was significantly increased by extracellular inositol and glucose, it was strongly impaired by SLC transporter inhibition. In the GBM cell monolayers, both SLCs were exclusively detected in the migrating cells at the monolayer edge. In single GBM cells, both transporters were primarily localized at the leading edge of the lamellipodium. Interestingly, in GBM cells migrating via blebbing, SLC5A1 and SLC5A3 were predominantly detected in nascent and mature blebs, respectively. (4) Conclusion: We provide several lines of evidence for the involvement of SLC5A1 and SLC5A3 in GBM cell migration, thereby complementing the migration-associated transportome. Our findings suggest that SLC inhibition is a promising approach to GBM treatment.
Collapse
Affiliation(s)
- Philippa K. Brosch
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Tessa Korsa
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
| | - Danush Taban
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Patrick Eiring
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Philipp Kreisz
- Julius-von-Sachs Institute, University of Würzburg, 97082 Würzburg, Germany;
| | - Sascha Hildebrand
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Julia Neubauer
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
- Department of Molecular and Cellular Biotechnology, Saarland University, 66123 Saarbrücken, Germany
- Faculty of Marine Science, Universidad Católica del Norte, Coquimbo 1281, Chile
| | - Markus Sauer
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Ryo Shirakashi
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan;
| | - Cholpon S. Djuzenova
- Department of Radiation Oncology, University Hospital of Würzburg, 97080 Würzburg, Germany;
| | - Dmitri Sisario
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Vladimir L. Sukhorukov
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| |
Collapse
|
15
|
Pittman M, Iu E, Li K, Wang M, Chen J, Taneja N, Jo MH, Park S, Jung WH, Liang L, Barman I, Ha T, Gaitanaros S, Liu J, Burnette D, Plotnikov S, Chen Y. Membrane Ruffling is a Mechanosensor of Extracellular Fluid Viscosity. NATURE PHYSICS 2022; 18:1112-1121. [PMID: 37220497 PMCID: PMC10202009 DOI: 10.1038/s41567-022-01676-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/17/2022] [Indexed: 05/25/2023]
Abstract
Cell behaviour is affected by the physical forces and mechanical properties of the cells and of their microenvironment. The viscosity of extracellular fluid - a component of the cellular microenvironment - can vary by orders of magnitude, but its effect on cell behaviour remains largely unexplored. Using bio-compatible polymers to increase the viscosity of the culture medium, we characterize how viscosity affects cell behaviour. We find that multiple types of adherent cells respond in an unexpected but similar manner to elevated viscosity. In a highly viscous medium, cells double their spread area, exhibit increased focal adhesion formation and turnover, generate significantly greater traction forces, and migrate nearly two times faster. We observe that when cells are immersed in regular medium, these viscosity-dependent responses require an actively ruffling lamellipodium - a dynamic membrane structure at the front of the cell. We present evidence that cells utilize membrane ruffling to sense changes in extracellular fluid viscosity and to trigger adaptive responses.
Collapse
Affiliation(s)
- Matthew Pittman
- Department of Mechanical Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Center for Cell Dynamics, Johns Hopkins University
| | - Ernest Iu
- Department of Cell & Systems Biology, University of Toronto
| | - Keva Li
- Department of Mechanical Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Center for Cell Dynamics, Johns Hopkins University
| | - Mingjiu Wang
- Department of Mechanical Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Center for Cell Dynamics, Johns Hopkins University
| | - Junjie Chen
- Department of Mechanical Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Center for Cell Dynamics, Johns Hopkins University
| | - Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University
| | | | - Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Center for Cell Dynamics, Johns Hopkins University
| | - Wei-Hung Jung
- Department of Mechanical Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Center for Cell Dynamics, Johns Hopkins University
| | - Le Liang
- Department of Mechanical Engineering, Johns Hopkins University
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University
| | - Taekjip Ha
- Department of Biophysics, Johns Hopkins University
| | | | - Jian Liu
- Department of Cell Biology, Johns Hopkins University School of Medicine
| | - Dylan Burnette
- Department of Cell and Developmental Biology, Vanderbilt University
| | | | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Center for Cell Dynamics, Johns Hopkins University
| |
Collapse
|
16
|
Molina E, Cataldo VF, Eggers C, Muñoz-Madrid V, Glavic Á. p53 Related Protein Kinase is Required for Arp2/3-Dependent Actin Dynamics of Hemocytes in Drosophila melanogaster. Front Cell Dev Biol 2022; 10:859105. [PMID: 35721516 PMCID: PMC9201722 DOI: 10.3389/fcell.2022.859105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022] Open
Abstract
Cells extend membrane protrusions like lamellipodia and filopodia from the leading edge to sense, to move and to form new contacts. The Arp2/3 complex sustains lamellipodia formation, and in conjunction with the actomyosin contractile system, provides mechanical strength to the cell. Drosophila p53-related protein kinase (Prpk), a Tsc5p ortholog, has been described as essential for cell growth and proliferation. In addition, Prpk interacts with proteins associated to actin filament dynamics such as α-spectrin and the Arp2/3 complex subunit Arpc4. Here, we investigated the role of Prpk in cell shape changes, specifically regarding actin filament dynamics and membrane protrusion formation. We found that reductions in Prpk alter cell shape and the structure of lamellipodia, mimicking the phenotypes evoked by Arp2/3 complex deficiencies. Prpk co-localize and co-immunoprecipitates with the Arp2/3 complex subunit Arpc1 and with the small GTPase Rab35. Importantly, expression of Rab35, known by its ability to recruit upstream regulators of the Arp2/3 complex, could rescue the Prpk knockdown phenotypes. Finally, we evaluated the requirement of Prpk in different developmental contexts, where it was shown to be essential for correct Arp2/3 complex distribution and actin dynamics required for hemocytes migration, recruitment, and phagocytosis during immune response.
Collapse
Affiliation(s)
- Emiliano Molina
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Vicente F. Cataldo
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Eggers
- Department for Chemistry and Biochemistry and Pharmaceutical Sciences, Faculty of Science, University of Bern, Bern, Switzerland
| | - Valentina Muñoz-Madrid
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Álvaro Glavic
- FONDAP Center for Genome Regulation, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- *Correspondence: Álvaro Glavic,
| |
Collapse
|
17
|
Powers RM, Daza R, Koehler AE, Courchet J, Calabrese B, Hevner RF, Halpain S. Growth cone macropinocytosis of neurotrophin receptor and neuritogenesis are regulated by neuron navigator 1. Mol Biol Cell 2022; 33:ar64. [PMID: 35352947 PMCID: PMC9561856 DOI: 10.1091/mbc.e21-12-0623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neuron navigator 1 (Nav1) is a cytoskeleton-associated protein expressed during brain development that is necessary for proper neuritogenesis, but the underlying mechanisms are poorly understood. Here we show that Nav1 is present in elongating axon tracts during mouse brain embryogenesis. We found that depletion of Nav1 in cultured neurons disrupts growth cone morphology and neurotrophin-stimulated neuritogenesis. In addition to regulating both F-actin and microtubule properties, Nav1 promotes actin-rich membrane ruffles in the growth cone and promotes macropinocytosis at those membrane ruffles, including internalization of the TrkB receptor for the neurotrophin brain-derived neurotropic factor (BDNF). Growth cone macropinocytosis is important for downstream signaling, neurite targeting, and membrane recycling, implicating Nav1 in one or more of these processes. Depletion of Nav1 also induces transient membrane blebbing via disruption of signaling in the Rho GTPase signaling pathway, supporting the novel role of Nav1 in dynamic actin-based membrane regulation at the cell periphery. These data demonstrate that Nav1 works at the interface of microtubules, actin, and plasma membrane to organize the cell periphery and promote uptake of growth and guidance cues to facilitate neural morphogenesis during development.
Collapse
Affiliation(s)
- Regina M. Powers
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037
| | - Ray Daza
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,Department of Pathology, University of California, San Diego, La Jolla, CA 92161
| | - Alanna E. Koehler
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,Department of Pathology, University of California, San Diego, La Jolla, CA 92161
| | - Julien Courchet
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon I, 69008 Lyon Cedex, France
| | - Barbara Calabrese
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037
| | - Robert F. Hevner
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,Department of Pathology, University of California, San Diego, La Jolla, CA 92161
| | - Shelley Halpain
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037,*Address correspondence to: Shelley Halpain ()
| |
Collapse
|
18
|
Chen Y, Zhai MJ, Mehwish N, Xu MD, Wang Y, Gong YX, Ren MM, Deng H, Lee BH. Comparison of globular albumin methacryloyl and random-coil gelatin methacryloyl: Preparation, hydrogel properties, cell behaviors, and mineralization. Int J Biol Macromol 2022; 204:692-708. [PMID: 35150780 DOI: 10.1016/j.ijbiomac.2022.02.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/28/2022] [Accepted: 02/07/2022] [Indexed: 12/19/2022]
Abstract
Bovine serum albumin methacryloyl (BSAMA) is a newly emerging photocurable globular protein-based material whereas gelatin methacryloyl (GelMA) is one of the most popular photocurable fibrous protein-based materials. So far, the influence of their different structural conformations as building blocks on hydrogel properties and mineral deposition has not been investigated. Here, we compared their differences in structures, gelation kinetics, hydrogel properties, mineralization, and cell behaviors. BSAMA maintained a stable globular structure while GelMA exhibited temperature-sensitive conformations (4 - 37 °C). BSAMA displayed slower gelation kinetics and much more retarded enzymatic degradation compared to GelMA. Photocurable BSAMA (6.41 - 390.95 kPa) and GelMA hydrogels (36.09 - 199.70 kPa) exhibited tunable mechanical properties depending on their concentrations (10 - 20%). Interestingly, BSAMA hydrogels mineralized needle-like apatite (Ca/P: 1.409) with higher crystallinity compared to GelMA hydrogels (Ca/P: 1.344). BSAMA and GelMA supported satisfactory cell (MC3T3-L1) viability of 99.43 ± 0.57% and 97.14 ± 0.69%, respectively. However, BSAMA gels were less favorable to cell proliferation and migration than GelMA gels. In serum-free environments, cells on GelMA displayed a higher amount of attachment, a more elongated shape, and a longer protrusion compared to those on BSAMA (p < 0.01) during the early adhesion.
Collapse
Affiliation(s)
- Yuan Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China; Department of Periodontics, School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Meng Jiao Zhai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Nabila Mehwish
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Meng Die Xu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Yi Wang
- Department of Orthodontics, School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yi Xuan Gong
- Department of Periodontics, School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Man Man Ren
- Department of Periodontics, School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Hui Deng
- Department of Periodontics, School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Bae Hoon Lee
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China; Oujiang Laboratory (Zhejiang Lab for Rengerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China.
| |
Collapse
|
19
|
Zhao X, Fisher ES, Wang Y, Zuloaga K, Manley L, Temple S. 4D imaging analysis of the aging mouse neural stem cell niche reveals a dramatic loss of progenitor cell dynamism regulated by the RHO-ROCK pathway. Stem Cell Reports 2022; 17:245-258. [PMID: 35030320 PMCID: PMC8828534 DOI: 10.1016/j.stemcr.2021.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/02/2022] Open
Abstract
In the adult ventricular-subventricular zone (V-SVZ), neural stem cells (NSCs) give rise to transit-amplifying progenitor (TAP) cells. These progenitors reside in different subniche locations, implying that cell movement must accompany lineage progression, but the dynamic behaviors of adult NSCs and TAPs remain largely unexplored. Here, we performed live time-lapse imaging with computer-based image analysis of young and aged 3D V-SVZ wholemounts from transgenic mice with fluorescently distinguished NSCs and TAP cells. Young V-SVZ progenitors are highly dynamic, with regular process outgrowth and retraction and cell migration. However, these activities dramatically declined with age. An examination of single-cell RNA sequencing (RNA-seq) data revealed age-associated changes in the Rho-Rock pathway that are important for cell motility. Applying a small molecule to inhibit ROCK transformed young into old V-SVZ progenitor cell dynamic behaviors. Hence RHO-ROCK signaling is critical for normal adult NSC and TAP movement and interactions, which are compromised with age, concomitant with the loss of regenerative ability.
Collapse
Affiliation(s)
- Xiuli Zhao
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | | | - Yue Wang
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Kristen Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical Center, Albany NY 12208, USA
| | - Luke Manley
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
20
|
Berlew EE, Yamada K, Kuznetsov IA, Rand EA, Ochs CC, Jaber Z, Gardner KH, Chow BY. Designing Single-Component Optogenetic Membrane Recruitment Systems: The Rho-Family GTPase Signaling Toolbox. ACS Synth Biol 2022; 11:515-521. [PMID: 34978789 PMCID: PMC8867532 DOI: 10.1021/acssynbio.1c00604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We describe the efficient creation of single-component optogenetic tools for membrane recruitment-based signaling perturbation using BcLOV4 technology. The workflow requires two plasmids to create six different domain arrangements of the dynamic membrane binder BcLOV4, a fluorescent reporter, and the fused signaling protein of interest. Screening of this limited set of genetic constructs for expression characteristics and dynamic translocation in response to one pulse of light is sufficient to identify viable signaling control tools. The reliability of this streamlined approach is demonstrated by the creation of an optogenetic Cdc42 GTPase and Rac1-activating Tiam1 GEF protein, which together with our other recently reported technologies, completes a toolbox for spatiotemporally precise induction of Rho-family GTPase signaling at the GEF or GTPase level, for driving filopodial protrusions, lamellipodial protrusions, and cell contractility, respectively mediated by Cdc42, Rac1, and RhoA.
Collapse
Affiliation(s)
- Erin E. Berlew
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104, USA
| | - Keisuke Yamada
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104, USA,Department of Electrical Engineering and Bioscience, Faculty of Science and Engineering, Waseda University, Tokyo, Japan 169-8050
| | - Ivan A. Kuznetsov
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104, USA
| | - Eleanor A. Rand
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104, USA,Department of Systems Biology, Harvard University Medical School, Boston MA 02115, USA
| | - Chandler C. Ochs
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104, USA,McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Zaynab Jaber
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031, USA,Ph.D. Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10016, USA
| | - Kevin H. Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031, USA,Department of Chemistry and Biochemistry, City College of New York, New York, NY 10031, USA,Ph.D. Programs in Biochemistry, Chemistry, and Biology, The Graduate Center, City University of New York, New York, NY 10016, USA
| | - Brian Y. Chow
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA 19104, USA,Correspondence: ; 210 S 33rd Street, Suite 240, Philadelphia, PA 19104; (+1) (215) 898-5159
| |
Collapse
|
21
|
Peng L, Gautrot JE. Long term expansion profile of mesenchymal stromal cells at protein nanosheet-stabilised bioemulsions for next generation cell culture microcarriers. Mater Today Bio 2021; 12:100159. [PMID: 34841241 PMCID: PMC8605361 DOI: 10.1016/j.mtbio.2021.100159] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 12/27/2022] Open
Abstract
Tremendous progress in the identification, isolation and expansion of stem cells has allowed their application in regenerative medicine and tissue engineering, and their use as advanced in vitro models. As a result, stem cell manufacturing increasingly requires scale up, parallelisation and automation. However, solid substrates currently used for the culture of adherent cells are poorly adapted for such applications, owing to their difficult processing from cell products, relatively high costs and their typical reliance on difficult to recycle plastics and microplastics. In this work, we show that bioemulsions formed of microdroplets stabilised by protein nanosheets displaying strong interfacial mechanics are well-suited for the scale up of adherent stem cells such as mesenchymal stromal cells (MSCs). We demonstrate that, over multiple passages (up to passage 10), MSCs retain comparable phenotypes when cultured on such bioemulsions, solid microcarriers (Synthemax II) and classic 2D tissue culture polystyrene. Phenotyping (cell proliferation, morphometry, flow cytometry and differentiation assays) of MSCs cultured for multiple passages on these systems indicate that, although stemness is lost at late passages when cultured on these different substrates, stem cell phenotypes remained comparable between different culture conditions, at any given passage. Hence our study validates the use of bioemulsions for the long term expansion of adherent stem cells and paves the way to the design of novel 3D bioreactors based on microdroplet microcarriers.
Collapse
Affiliation(s)
- Lihui Peng
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Julien E Gautrot
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| |
Collapse
|
22
|
Holt JR, Zeng WZ, Evans EL, Woo SH, Ma S, Abuwarda H, Loud M, Patapoutian A, Pathak MM. Spatiotemporal dynamics of PIEZO1 localization controls keratinocyte migration during wound healing. eLife 2021; 10:65415. [PMID: 34569935 PMCID: PMC8577841 DOI: 10.7554/elife.65415] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 09/24/2021] [Indexed: 12/17/2022] Open
Abstract
Keratinocytes, the predominant cell type of the epidermis, migrate to reinstate the epithelial barrier during wound healing. Mechanical cues are known to regulate keratinocyte re-epithelialization and wound healing; however, the underlying molecular transducers and biophysical mechanisms remain elusive. Here, we show through molecular, cellular, and organismal studies that the mechanically activated ion channel PIEZO1 regulates keratinocyte migration and wound healing. Epidermal-specific Piezo1 knockout mice exhibited faster wound closure while gain-of-function mice displayed slower wound closure compared to littermate controls. By imaging the spatiotemporal localization dynamics of endogenous PIEZO1 channels, we find that channel enrichment at some regions of the wound edge induces a localized cellular retraction that slows keratinocyte collective migration. In migrating single keratinocytes, PIEZO1 is enriched at the rear of the cell, where maximal retraction occurs, and we find that chemical activation of PIEZO1 enhances retraction during single as well as collective migration. Our findings uncover novel molecular mechanisms underlying single and collective keratinocyte migration that may suggest a potential pharmacological target for wound treatment. More broadly, we show that nanoscale spatiotemporal dynamics of Piezo1 channels can control tissue-scale events, a finding with implications beyond wound healing to processes as diverse as development, homeostasis, disease, and repair.
Collapse
Affiliation(s)
- Jesse R Holt
- Departmentof Physiology & Biophysics, UC Irvine, Irvine, United States.,Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, United States.,Center for Complex Biological Systems, UC Irvine, Irvine, United States
| | - Wei-Zheng Zeng
- Howard Hughes Medical Institute, Department of Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Elizabeth L Evans
- Departmentof Physiology & Biophysics, UC Irvine, Irvine, United States.,Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, United States
| | - Seung-Hyun Woo
- Howard Hughes Medical Institute, Department of Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Shang Ma
- Howard Hughes Medical Institute, Department of Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Hamid Abuwarda
- Departmentof Physiology & Biophysics, UC Irvine, Irvine, United States.,Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, United States
| | - Meaghan Loud
- Howard Hughes Medical Institute, Department of Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Ardem Patapoutian
- Howard Hughes Medical Institute, Department of Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Medha M Pathak
- Departmentof Physiology & Biophysics, UC Irvine, Irvine, United States.,Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, United States.,Center for Complex Biological Systems, UC Irvine, Irvine, United States.,Department of Biomedical Engineering, UC Irvine, Irvine, United States
| |
Collapse
|
23
|
Porro C, Pennella A, Panaro MA, Trotta T. Functional Role of Non-Muscle Myosin II in Microglia: An Updated Review. Int J Mol Sci 2021; 22:ijms22136687. [PMID: 34206505 PMCID: PMC8267657 DOI: 10.3390/ijms22136687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Myosins are a remarkable superfamily of actin-based motor proteins that use the energy derived from ATP hydrolysis to translocate actin filaments and to produce force. Myosins are abundant in different types of tissues and involved in a large variety of cellular functions. Several classes of the myosin superfamily are expressed in the nervous system; among them, non-muscle myosin II (NM II) is expressed in both neurons and non-neuronal brain cells, such as astrocytes, oligodendrocytes, endothelial cells, and microglia. In the nervous system, NM II modulates a variety of functions, such as vesicle transport, phagocytosis, cell migration, cell adhesion and morphology, secretion, transcription, and cytokinesis, as well as playing key roles during brain development, inflammation, repair, and myelination functions. In this review, we will provide a brief overview of recent emerging roles of NM II in resting and activated microglia cells, the principal regulators of immune processes in the central nervous system (CNS) in both physiological and pathological conditions. When stimulated, microglial cells react and produce a number of mediators, such as pro-inflammatory cytokines, free radicals, and nitric oxide, that enhance inflammation and contribute to neurodegenerative diseases. Inhibition of NM II could be a new therapeutic target to treat or to prevent CNS diseases.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Antonio Pennella
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
- Correspondence:
| |
Collapse
|
24
|
Jaafar IH, Jedlicka SS, Coulter JP. Poly(glycerol sebacate) – a revolutionary biopolymer. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2020-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Abstract
Novel materials possessing physical, mechanical, and chemical properties similar to those found in vivo provide a potential platform for building artificial microenvironments for tissue engineering applications. Poly(glycerol sebacate) is one such material. It has tunable mechanical properties within the range of common tissue, and favorable cell response without surface modification with adhesive ligands, and biodegradability. In this chapter, an overview of the material is presented, focusing on synthesis, characterization, microfabrication, use as a substrate in in vitro mammalian cell culture, and degradation characteristics.
Collapse
Affiliation(s)
- Israd H. Jaafar
- Mechanical Engineering , Utah Valley University , 800 W University Parkway , Orem , UT , USA
| | - Sabrina S. Jedlicka
- Materials Science and Engineering , Lehigh University , Bethlehem , PA , USA
| | - John P. Coulter
- Mechanical Engineering and Mechanics , Lehigh University , Bethlehem , PA , USA
| |
Collapse
|
25
|
Voelkner C, Wendt M, Lange R, Ulbrich M, Gruening M, Staehlke S, Nebe B, Barke I, Speller S. The nanomorphology of cell surfaces of adhered osteoblasts. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:242-256. [PMID: 33777612 PMCID: PMC7961864 DOI: 10.3762/bjnano.12.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
The functionality of living cells is inherently linked to subunits with dimensions ranging from several micrometers down to the nanometer scale. The cell surface plays a particularly important role. Electric signaling, including information processing, takes place at the membrane, as well as adhesion and contact. For osteoblasts, adhesion and spreading are crucial processes with regard to bone implants. Here we present a comprehensive characterization of the 3D nanomorphology of living, as well as fixed, osteoblastic cells using scanning ion conductance microscopy (SICM), which is a nanoprobing method that largely avoids mechanical perturbations. Dynamic ruffles are observed, manifesting themselves in characteristic membrane protrusions. They contribute to the overall surface corrugation, which we systematically study by introducing the relative 3D excess area as a function of the projected adhesion area. A clear anticorrelation between the two parameters is found upon analysis of ca. 40 different cells on glass and on amine-covered surfaces. At the rim of lamellipodia, characteristic edge heights between 100 and 300 nm are observed. Power spectral densities of membrane fluctuations show frequency-dependent decay exponents with absolute values greater than 2 on living osteoblasts. We discuss the capability of apical membrane features and fluctuation dynamics in aiding the assessment of adhesion and migration properties on a single-cell basis.
Collapse
Affiliation(s)
- Christian Voelkner
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
- Institute of Physics, University of Rostock, Albert-Einstein-Str. 23, 18059 Rostock, Germany
| | - Mirco Wendt
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
- Institute of Physics, University of Rostock, Albert-Einstein-Str. 23, 18059 Rostock, Germany
| | - Regina Lange
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
- Institute of Physics, University of Rostock, Albert-Einstein-Str. 23, 18059 Rostock, Germany
| | - Max Ulbrich
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
- Institute of Physics, University of Rostock, Albert-Einstein-Str. 23, 18059 Rostock, Germany
| | - Martina Gruening
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany
| | - Susanne Staehlke
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany
| | - Barbara Nebe
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany
| | - Ingo Barke
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
- Institute of Physics, University of Rostock, Albert-Einstein-Str. 23, 18059 Rostock, Germany
| | - Sylvia Speller
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
- Institute of Physics, University of Rostock, Albert-Einstein-Str. 23, 18059 Rostock, Germany
| |
Collapse
|
26
|
Liu WQ, Li WL, Ma SM, Liang L, Kou ZY, Yang J. Discovery of core gene families associated with liver metastasis in colorectal cancer and regulatory roles in tumor cell immune infiltration. Transl Oncol 2021; 14:101011. [PMID: 33450702 PMCID: PMC7810789 DOI: 10.1016/j.tranon.2021.101011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 01/21/2023] Open
Abstract
In this study, we aimed to uncover genes that drive the pathogenesis of liver metastasis in colorectal cancer (CRC), and identify effective genes that could serve as potential therapeutic targets for treating with colorectal liver metastasis patients based on two GEO datasets. Several bioinformatics approaches were implemented. First, differential expression analysis screened out key differentially expressed genes (DEGs) across the two GEO datasets. Based on gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, we identified the enrichment functions and pathways of the DEGs that were associated with liver metastasis in CRC. Second, immune infiltration analysis identified key immune signature gene sets associated with CRC liver metastasis, among which two key immune gene families (CD and CCL) identified as key DEGs were filtered by protein-protein interaction (PPI) network. Some of the members in these gene families were associated with disease free survival (DFS) or overall survival (OS) in two subtypes of CRC, namely COAD and READ. Finally, functional enrichment analysis of the two gene families and their neighboring genes revealed that they were closely associated with cytokine, leukocyte proliferation and chemotaxis. These results are valuable in comprehending the pathogenesis of liver metastasis in CRC, and are of seminal importance in understanding the role of immune tumor infiltration in CRC. Our study also identified potentially effective therapeutic targets for liver metastasis in CRC including CCL20, CCL24 and CD70.
Collapse
Affiliation(s)
- Wei-Qing Liu
- Department of Internal Medicine-Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, PR China
| | - Wen-Liang Li
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang road, Kunming, Yunnan 650032, PR China
| | - Shu-Min Ma
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang road, Kunming, Yunnan 650032, PR China
| | - Lei Liang
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang road, Kunming, Yunnan 650032, PR China
| | - Zhi-Yong Kou
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang road, Kunming, Yunnan 650032, PR China
| | - Jun Yang
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295 Xichang road, Kunming, Yunnan 650032, PR China.
| |
Collapse
|
27
|
Zheng Y, Montague SJ, Lim YJ, Xu T, Xu T, Gardiner EE, Lee WM. Label-free multimodal quantitative imaging flow assay for intrathrombus formation in vitro. Biophys J 2021; 120:791-804. [PMID: 33513336 DOI: 10.1016/j.bpj.2021.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/17/2020] [Accepted: 01/13/2021] [Indexed: 10/22/2022] Open
Abstract
Microfluidics in vitro assays recapitulate a blood vessel microenvironment using surface-immobilized agonists under biofluidic flows. However, these assays do not quantify intrathrombus mass and activities of adhesive platelets at the agonist margin and use fluorescence labeling, therefore limiting clinical translation potential. Here, we describe a label-free multimodal quantitative imaging flow assay that combines rotating optical coherent scattering microscopy and quantitative phase microscopy. The combined imaging platform enables real-time evaluation of membrane fluctuations of adhesive-only platelets and total intrathrombus mass under physiological flow rates in vitro. We call this multimodal quantitative imaging flow assay coherent optical scattering and phase interferometry (COSI). COSI records intrathrombus mass to picogram accuracy and shape changes to a platelet membrane with high spatial-temporal resolution (0.4 μm/s) under physiological and pathophysiological fluid shear stress (1800 and 7500 s-1). With COSI, we generate an axial slice of 4 μm from the coverslip surface, approximately equivalent to the thickness of a single platelet, which permits nanoscale quantification of membrane fluctuation (activity) of adhesive platelets during initial adhesion, spreading, and recruitment into a developing thrombus (mass). Under fluid shear, pretreatment with a broad range metalloproteinase inhibitor (250 μM GM6001) blocked shedding of platelet adhesion receptors that shown elevated adhesive platelet activity at average of 42.1 μm/s and minimal change in intrathrombus mass.
Collapse
Affiliation(s)
- Yujie Zheng
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Samantha J Montague
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Yean J Lim
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research; ACRF Centre for Intravital Imaging of Niches for Cancer Immune Therapy
| | - Tao Xu
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Tienan Xu
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Woei Ming Lee
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research; ACRF Centre for Intravital Imaging of Niches for Cancer Immune Therapy; The ARC Centre of Excellence in Advanced Molecular Imaging, The Australian National University, Canberra, Australia.
| |
Collapse
|
28
|
Schreiber C, Amiri B, Heyn JCJ, Rädler JO, Falcke M. On the adhesion-velocity relation and length adaptation of motile cells on stepped fibronectin lanes. Proc Natl Acad Sci U S A 2021; 118:e2009959118. [PMID: 33483418 PMCID: PMC7869109 DOI: 10.1073/pnas.2009959118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The biphasic adhesion-velocity relation is a universal observation in mesenchymal cell motility. It has been explained by adhesion-promoted forces pushing the front and resisting motion at the rear. Yet, there is little quantitative understanding of how these forces control cell velocity. We study motion of MDA-MB-231 cells on microlanes with fields of alternating Fibronectin densities to address this topic and derive a mathematical model from the leading-edge force balance and the force-dependent polymerization rate. It reproduces quantitatively our measured adhesion-velocity relation and results with keratocytes, PtK1 cells, and CHO cells. Our results confirm that the force pushing the leading-edge membrane drives lamellipodial retrograde flow. Forces resisting motion originate along the whole cell length. All motion-related forces are controlled by adhesion and velocity, which allows motion, even with higher Fibronectin density at the rear than at the front. We find the pathway from Fibronectin density to adhesion structures to involve strong positive feedbacks. Suppressing myosin activity reduces the positive feedback. At transitions between different Fibronectin densities, steady motion is perturbed and leads to changes of cell length and front and rear velocity. Cells exhibit an intrinsic length set by adhesion strength, which, together with the length dynamics, suggests a spring-like front-rear interaction force. We provide a quantitative mechanistic picture of the adhesion-velocity relation and cell response to adhesion changes integrating force-dependent polymerization, retrograde flow, positive feedback from integrin to adhesion structures, and spring-like front-rear interaction.
Collapse
Affiliation(s)
- Christoph Schreiber
- Faculty of Physics and Center for NanoScience, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| | - Behnam Amiri
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Johannes C J Heyn
- Faculty of Physics and Center for NanoScience, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| | - Joachim O Rädler
- Faculty of Physics and Center for NanoScience, Ludwig-Maximilians-Universität München, 80539 Munich, Germany;
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany;
- Department of Physics, Humboldt University, 12489 Berlin, Germany
| |
Collapse
|
29
|
The Cycling of Intracellular Calcium Released in Response to Fluid Shear Stress Is Critical for Migration-Associated Actin Reorganization in Eosinophils. Cells 2021; 10:cells10010157. [PMID: 33467432 PMCID: PMC7829934 DOI: 10.3390/cells10010157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 01/08/2023] Open
Abstract
The magnitude of eosinophil mobilization into respiratory tissues drives the severity of inflammation in several airway diseases. In classical models of leukocyte extravasation, surface integrins undergo conformational switches to high-affinity states via chemokine binding activation. Recently, we learned that eosinophil integrins possess mechanosensitive properties that detect fluid shear stress, which alone was sufficient to induce activation. This mechanical stimulus triggered intracellular calcium release and hallmark migration-associated cytoskeletal reorganization including flattening for increased cell–substratum contact area and pseudopodia formation. The present study utilized confocal fluorescence microscopy to investigate the effects of pharmacological inhibitors to calcium signaling and actin polymerization pathways on shear stress-induced migration in vitro. Morphological changes (cell elongation, membrane protrusions) succeeded the calcium flux in untreated eosinophils within 2 min, suggesting that calcium signaling was upstream of actin cytoskeleton rearrangement. The inhibition of ryanodine receptors and endomembrane Ca2+-ATPases corroborated this idea, indicated by a significant increase in time between the calcium spike and actin polymerization. The impact of the temporal link is evident as the capacity of treated eosinophils to move across fibronectin-coated surfaces was significantly hampered relative to untreated eosinophils. Furthermore, we determined that the nature of cellular motility in response to fluid shear stress was nondirectional.
Collapse
|
30
|
1'H-Indole-3'-Carbonyl-Thiazole-4-Carboxylic Acid Methyl Ester Blocked Human Glioma Cell Invasion via Aryl Hydrocarbon Receptor's Regulation of Cytoskeletal Contraction. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2616930. [PMID: 33083460 PMCID: PMC7556083 DOI: 10.1155/2020/2616930] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/10/2020] [Accepted: 08/24/2020] [Indexed: 11/17/2022]
Abstract
Blocking glioma cell invasion has been challenging due to cancer cells that can swiftly switch their migration mode, and agents that can block more than one migration mode are sought after. We found that small molecule 2-(1H-indole-3-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), an endogenous aryl hydrocarbon receptor (AHR) agonist, can block more than one mode of glioma cell migration, based on cultured cell behavior captured by videos. Data from wound-healing assays and mouse xenograft glioma models corroborated ITE's migration-inhibiting effects while knocking down AHR by siRNA abolished these effects. To identify genes that mediated ITE-AHR's effect, we first collected gene expression changes upon ITE treatment by RNA-seq, then compared them against literature reported migration-related genes in glioma and that were potentially regulated by AHR. MYH9, a component of nonmuscle myosin IIA (NMIIA), was confirmed to be reduced by ITE treatment. When MYH9 was overexpressed in the glioma cells, a good correlation was observed between the expression level and the cell migration ability, determined by wound-healing assay. Correspondingly, overexpression of MYH9 abrogated ITE's migration-inhibiting effects, indicating that ITE-AHR inhibited cell migration via inhibiting MYH9 expression. MYH9 is essential for cell migration in 3D confined space and not a discovered target of AHR; the fact that ITE affects MYH9 via AHR opens a new research and development avenue.
Collapse
|
31
|
Sobolewski C, Abegg D, Berthou F, Dolicka D, Calo N, Sempoux C, Fournier M, Maeder C, Ay AS, Clavien PA, Humar B, Dufour JF, Adibekian A, Foti M. S100A11/ANXA2 belongs to a tumour suppressor/oncogene network deregulated early with steatosis and involved in inflammation and hepatocellular carcinoma development. Gut 2020; 69:1841-1854. [PMID: 31919231 DOI: 10.1136/gutjnl-2019-319019] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) development occurs with non-alcoholic fatty liver disease (NAFLD) in the absence of cirrhosis and with an increasing incidence due to the obesity pandemic. Mutations of tumour suppressor (TS) genes and oncogenes (ONC) have been widely characterised in HCC. However, mounting evidence indicates that non-genomic alterations of TS/ONC occur early with NAFLD, thereby potentially promoting hepatocarcinogenesis in an inflammatory/fibrotic context. The aim of this study was to identify and characterise these alterations. DESIGN The proteome of steatotic liver tissues from mice spontaneously developing HCC was analysed. Alterations of TSs/ONCs were further investigated in various mouse models of NAFLD/HCC and in human samples. The inflammatory, fibrogenic and oncogenic functions of S100A11 were assessed through in vivo, in vitro and ex-vivo analyses. RESULTS A whole set of TSs/ONCs, respectively, downregulated or upregulated was uncovered in mice and human with NAFLD. Alterations of these TSs/ONCs were preserved or even exacerbated in HCC. Among them, overexpression of S100A11 was associated with high-grade HCC and poor prognosis. S100A11 downregulation in vivo significantly restrains the development of inflammation and fibrosis in mice fed a choline/methionine-deficient diet. Finally, in vitro and ex-vivo analyses revealed that S100A11 is a marker of hepatocyte de-differentiation, secreted by cancer cells, and promoting cell proliferation and migration. CONCLUSION Cellular stress associated with NAFLD triggers non-genomic alterations of a whole network of TSs/ONCs fostering hepatocarcinogenesis. Among those, overexpression of the oncogenic factor S100A11 promotes inflammation/fibrosis in vivo and is significantly associated with high-grade HCC with poor prognosis.
Collapse
Affiliation(s)
- Cyril Sobolewski
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| | - Daniel Abegg
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida, USA
| | - Flavien Berthou
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| | - Dobrochna Dolicka
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| | - Nicolas Calo
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| | - Christine Sempoux
- Department of Clinical Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Margot Fournier
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| | - Christine Maeder
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| | - Anne-Sophie Ay
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| | - Pierre-Alain Clavien
- Visceral and Transplantation Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Bostjan Humar
- Department of Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jean-François Dufour
- Department of Hepatology and Clinical Research, University of Bern, Bern, Switzerland
| | - Alexander Adibekian
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida, USA
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneve, GE, Switzerland
| |
Collapse
|
32
|
Gheorghe RO, Deftu A, Filippi A, Grosu A, Bica-Popi M, Chiritoiu M, Chiritoiu G, Munteanu C, Silvestro L, Ristoiu V. Silencing the Cytoskeleton Protein Iba1 (Ionized Calcium Binding Adapter Protein 1) Interferes with BV2 Microglia Functioning. Cell Mol Neurobiol 2020; 40:1011-1027. [PMID: 31950314 PMCID: PMC11448859 DOI: 10.1007/s10571-020-00790-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/07/2020] [Indexed: 01/31/2023]
Abstract
Iba1 (ionized calcium binding adapter protein 1) is a cytoskeleton protein specific only for microglia and macrophages, where it acts as an actin-cross linking protein. Although frequently regarded as a marker of activation, its involvement in cell migration, membrane ruffling, phagocytosis or in microglia remodeling during immunological surveillance of the brain suggest that Iba1 is not a simple cytoskeleton protein, but a signaling molecule involved in specific signaling pathways. In this study we investigated if Iba1 could also represent a drug target, and tested the hypothesis that its specific silencing with customized Iba1-siRNA can modulate microglia functioning. The results showed that Iba1-silenced BV2 microglia migrate less due to reduced proliferation and cell adhesion, while their phagocytic activity and P2x7 functioning was significantly increased. Our data are the proof of concept that Iba1 protein is a new microglia target, which opens a new therapeutic avenue for modulating microglia behavior.
Collapse
Affiliation(s)
- Roxana-Olimpia Gheorghe
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Alexandru Deftu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Alexandru Filippi
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Andreea Grosu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Melania Bica-Popi
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Marioara Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Gabriela Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Cristian Munteanu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Luigi Silvestro
- Pharma Serv International, Sabinelor 52, 050853, Bucharest, Romania
| | - Violeta Ristoiu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania.
| |
Collapse
|
33
|
Bilodeau P, Jacobsen D, Law-Vinh D, Lee JM. Phosphatidylinositol 4-kinase III beta regulates cell shape, migration, and focal adhesion number. Mol Biol Cell 2020; 31:1904-1916. [PMID: 32583740 PMCID: PMC7525810 DOI: 10.1091/mbc.e19-11-0600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Cell shape is regulated by cell adhesion and cytoskeletal and membrane dynamics. Cell shape, adhesion, and motility have a complex relationship and understanding them is important in understanding developmental patterning and embryogenesis. Here we show that the lipid kinase phosphatidylinositol 4-kinase III beta (PI4KIIIβ) regulates cell shape, migration, and focal adhesion (FA) number. PI4KIIIβ generates phosphatidylinositol 4-phosphate (PI4P) from phosphatidylinositol and is highly expressed in a subset of human breast cancers. PI4KIIIβ and the PI4P it generates regulate a variety of cellular functions, ranging from control of Golgi structure, fly fertility, and Akt signaling. Here, we show that loss of PI4KIIIβ expression decreases cell migration and alters cell shape in NIH3T3 fibroblasts. The changes are accompanied by an increase in the number of FA in cells lacking PI4KIIIβ. Furthermore, we find that PI4P-containing vesicles move to the migratory leading edge during migration and that some of these vesicles tether to and fuse with FA. Fusion is associated with FA disassembly. This suggests a novel regulatory role for PI4KIIIβ and PI4P in cell adhesion and cell shape maintenance.
Collapse
Affiliation(s)
- Patricia Bilodeau
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daniel Jacobsen
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Denise Law-Vinh
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jonathan M Lee
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
34
|
Xulu K, Duarte R, Augustine T. Combined Anastrozole and Antiplatelet Therapy Treatment Differentially Promotes Breast Cancer Cell Survival. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:497-508. [PMID: 32241309 DOI: 10.1017/s1431927620001324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Thromboembolic disorders are the second leading cause of death in breast cancer. Antiplatelet therapy combined with cancer therapy is a potential treatment strategy against cancer-associated thromboembolic disorders; however, the efficacy of such dual treatment has not been established. This study reports novel findings on the response of hormone-dependent breast cancer cell lines (MCF7/T47D) following 24 h treatment with Anastrozole, combined with Aspirin and Clopidogrel cocktail; and Atopaxar. Neutral red and lactate dehydrogenase assays were conducted to assess viability and cytotoxicity respectively. Flow cytometric Annexin-V/PI assay was used to assess the mode of cell death. Morphological alterations were studied using scanning electron microscopy. Statistical analysis was conducted using Statistica V13. Definitive outcomes were established with flow cytometric detection of phosphatidylserine exposure and propidium iodide staining, complemented with ultrastructural analysis. Results showed that a few cells were undergoing death mainly through secondary necrosis. Morphological features suggesting induced cell motility (pseudopodia/ruffled membranes) were observed in both cell lines; notably, T47D cells presented pronounced features than MCF7 cells. Overall, these findings suggest that such combined treatment may differentially promote cell survival, inducing a more aggressive breast cancer phenotype.
Collapse
Affiliation(s)
- Kutlwano Xulu
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193Johannesburg, South Africa
| | - Raquel Duarte
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193Johannesburg, South Africa
| | - Tanya Augustine
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193Johannesburg, South Africa
| |
Collapse
|
35
|
Hurley SK, Cutrone NM, Fath KR, Pajovich HT, Garcia J, Smith AM, Banerjee IA. Self-assembled phenylisoxazole-peptide hybrid assemblies and their interactions with breast and ovarian tumor cells. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2018.1525542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Sara K. Hurley
- Department of Chemistry, Fordham University, Bronx, NY, USA
| | | | - Karl R. Fath
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | - Jeremy Garcia
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | | |
Collapse
|
36
|
Metsiou DN, Siatis KE, Giannopoulou E, Papachristou DJ, Kalofonos HP, Koutras A, Athanassiou G. The Impact of Anti-tumor Agents on ER-Positive MCF-7 and HER2-Positive SKBR-3 Breast Cancer Cells Biomechanics. Ann Biomed Eng 2019; 47:1711-1724. [DOI: 10.1007/s10439-019-02284-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/02/2019] [Indexed: 01/07/2023]
|
37
|
Akishiba M, Futaki S. Inducible Membrane Permeabilization by Attenuated Lytic Peptides: A New Concept for Accessing Cell Interiors through Ruffled Membranes. Mol Pharm 2019; 16:2540-2548. [DOI: 10.1021/acs.molpharmaceut.9b00156] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Misao Akishiba
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
38
|
Murakami A, Maekawa M, Kawai K, Nakayama J, Araki N, Semba K, Taguchi T, Kamei Y, Takada Y, Higashiyama S. Cullin-3/KCTD10 E3 complex is essential for Rac1 activation through RhoB degradation in human epidermal growth factor receptor 2-positive breast cancer cells. Cancer Sci 2019; 110:650-661. [PMID: 30515933 PMCID: PMC6361568 DOI: 10.1111/cas.13899] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 01/01/2023] Open
Abstract
Rho GTPase Rac1 is a central regulator of F‐actin organization and signal transduction to control plasma membrane dynamics and cell proliferation. Dysregulated Rac1 activity is often observed in various cancers including breast cancer and is suggested to be critical for malignancy. Here, we showed that the ubiquitin E3 ligase complex Cullin‐3 (CUL3)/KCTD10 is essential for epidermal growth factor (EGF)‐induced/human epidermal growth factor receptor 2 (HER2)‐dependent Rac1 activation in HER2‐positive breast cancer cells. EGF‐induced dorsal membrane ruffle formation and cell proliferation that depends on both Rac1 and HER2 were suppressed in CUL3‐ or KCTD10‐depleted cells. Mechanistically, CUL3/KCTD10 ubiquitinated RhoB for degradation, another Rho GTPase that inhibits Rac1 activation at the plasma membrane by suppressing endosome‐to‐plasma membrane traffic of Rac1. In HER2‐positive breast cancers, high expression of Rac1 mRNA significantly correlated with poor prognosis of the patients. This study shows that this novel molecular axis (CUL3/KCTD10/RhoB) positively regulates the activity of Rac1 in HER2‐positive breast cancers, and our findings may lead to new treatment options for HER2‐ and Rac1‐positive breast cancers.
Collapse
Affiliation(s)
- Akari Murakami
- Department of Hepato-Biliary-Pancreatic Surgery and Breast Surgery, Ehime University Graduate School of Medicine, Toon, Japan.,Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan
| | - Masashi Maekawa
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan.,Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| | - Katsuhisa Kawai
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Japan
| | - Jun Nakayama
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Japan
| | - Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Japan
| | - Tomohiko Taguchi
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Yoshiaki Kamei
- Department of Hepato-Biliary-Pancreatic Surgery and Breast Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yasutsugu Takada
- Department of Hepato-Biliary-Pancreatic Surgery and Breast Surgery, Ehime University Graduate School of Medicine, Toon, Japan
| | - Shigeki Higashiyama
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Japan.,Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| |
Collapse
|
39
|
Kapperman HE, Goyeneche AA, Telleria CM. Mifepristone inhibits non-small cell lung carcinoma cellular escape from DNA damaging cisplatin. Cancer Cell Int 2018; 18:185. [PMID: 30479564 PMCID: PMC6238342 DOI: 10.1186/s12935-018-0683-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/09/2018] [Indexed: 12/23/2022] Open
Abstract
Background Lung cancer is the leading cause of cancer deaths in the world. The major histopathological subtype of lung cancer is non-small cell lung cancer (NSCLC). Platinum-based therapy is the standard of care for patients with advanced stage NSCLC. However, even with treatment, most patients will die of this disease within 5 years and most of these deaths are due to recurrence. One strategy to inhibit recurrence is to use cytostatic compounds following courses of lethal chemotherapy. We have shown in various cancer cell types that mifepristone (MF), an anti-progestin/anti-glucocorticoid, is a powerful cytostatic anti-cancer agent. Thus, in this work we tested the hypothesis that MF should be efficacious in inducing cytostasis and preventing repopulation of NSCLC following cisplatin (CDDP) therapy. Methods We established an in vitro approach wherein human NSCLC cells with different genetic backgrounds and sensitivities to CDDP (A549 and H23) were exposed to rounds of lethal concentrations of CDDP for 1 h followed or not by MF monotherapy. Every 2 days, cell number, cell viability, and colony-forming ability of viable cells were studied. Results CDDP killed the majority of cells, yet there were remnant cells escaping CDDP lethality and repopulating the culture, as evidenced by the improved clonogenic survival of viable cells. In contrast, when cells exposed to CDDP where further treated with MF following CDDP removal, their number and clonogenic capacity were reduced drastically. Conclusion This study reports that there is repopulation of NSCLC cells following a lethal concentration of CDDP monotherapy, that NSCLC cells are sensitive to the growth inhibition properties of MF, and that MF abrogates the repopulation of NSCLC cells following CDDP therapy. Our study supports further evaluating MF as an adjuvant therapy for NSCLC.
Collapse
Affiliation(s)
- Heather E Kapperman
- 2Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069 USA.,3Present Address: Eisenhower Army Medical Center, Ft. Gordon, GA USA
| | - Alicia A Goyeneche
- 1Experimental Pathology Unit, Department of Pathology, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC H3A 2B4 Canada
| | - Carlos M Telleria
- 1Experimental Pathology Unit, Department of Pathology, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC H3A 2B4 Canada
| |
Collapse
|
40
|
Bi J, Wang R, Zeng X. Lipid rafts regulate the lamellipodia formation of melanoma A375 cells via actin cytoskeleton-mediated recruitment of β1 and β3 integrin. Oncol Lett 2018; 16:6540-6546. [PMID: 30405793 PMCID: PMC6202517 DOI: 10.3892/ol.2018.9466] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/16/2018] [Indexed: 01/08/2023] Open
Abstract
Lipid rafts, distinct liquid-ordered plasma membrane microdomains, have been shown to regulate tumor cell migration by internalizing and recycling cell-surface proteins. The present study reports that lipid rafts are a prerequisite for lamellipodia formation, which is the first step in the processes of tumor cell migration. The results from the wound-healing assay and immunostaining indicated that lipid rafts were asymmetrically distributed to the leading edge of migrating melanoma A375 cells during lamellipodia formation. When the integrity of lipids rafts was disrupted, lamellipodia formation was inhibited. The investigation of possible molecular mechanisms indicated that lipid rafts recruited β1 and β3 integrins, two important adhesion proteins for cell migration, to the lamellipodia. However, the different distribution characteristics of β1 and β3 integrins implied disparate functions in lamellipodia formation. Further immunostaining experiments showed that the actin cytoskeleton was responsible for lipid raft-mediated β1 and β3 integrin distribution in the lamellipodia. Together, these findings provide novel insights into the regulation of lipid rafts in lamellipodia formation, and suggest that lipid rafts may be novel and attractive targets for cancer therapy.
Collapse
Affiliation(s)
- Jiajia Bi
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Ruifei Wang
- Key Laboratory for Microorganisms and Functional Molecules, College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, P.R. China
| | - Xianlu Zeng
- Institute of Genetics and Cytology, School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| |
Collapse
|
41
|
The role of SerpinB2 in human bronchial epithelial cells responses to particulate matter exposure. Arch Toxicol 2018; 92:2923-2933. [DOI: 10.1007/s00204-018-2259-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022]
|
42
|
Innocenti M. New insights into the formation and the function of lamellipodia and ruffles in mesenchymal cell migration. Cell Adh Migr 2018. [PMID: 29513145 DOI: 10.1080/19336918.2018.1448352] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Lamellipodia and ruffles are veil-shaped cell protrusions composed of a highly branched actin filament meshwork assembled by the Arp2/3 complex. These structures not only hallmark the leading edge of cells adopting the adhesion-based mesenchymal mode of migration but are also thought to drive cell movement. Although regarded as textbook knowledge, the mechanism of formation of lamellipodia and ruffles has been revisited in the last years leveraging new technologies. Furthermore, recent observations have also challenged our current view of the function of lamellipodia and ruffles in mesenchymal cell migration. Here, I review this literature and compare it with older studies to highlight the controversies and the outstanding open issues in the field. Moreover, I outline simple and plausible explanations to reconcile conflicting results and conclusions. Finally, I integrate the mechanisms regulating actin-based protrusion in a unifying model that accounts for random and ballistic mesenchymal cell migration.
Collapse
Affiliation(s)
- Metello Innocenti
- a Division of Molecular Genetics, The Netherlands Cancer Institute , Plesmanlaan 121, Amsterdam , CX , The Netherlands
| |
Collapse
|
43
|
Lamellipodial wrinkles in fish keratocytes as markers of imperfect coordination between extension and retraction during cell migration. Biochem Biophys Res Commun 2018. [PMID: 29530529 DOI: 10.1016/j.bbrc.2018.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell migration involves the precise coordination between extension at the front of the cell and retraction at the rear. This coordination is particularly evident in fast moving cells such as fish keratocytes, where it leads to highly stable gliding motion, propelled at the front by broad, 0.1-0-2 μm thick lamellipodia. Transient uncoupling between extension and retraction can occur if the rear is temporarily stuck, which might eventually lead to cell shape instabilities. We have frequently observed in fish keratocytes the presence of lamellipodial radial wrinkles, detected by confocal, scanning electron and side-view microscopy as folds in the lamellipodium up to 2 μm in height. Using a linear finite elements analysis, we simulated the displacement of cells either with perfect coordination between extension and retraction or with the rear transiently stuck while the front continues extending, and we observed that in this last condition compression stresses arise in the lamellipodium which predict the formation of the observed pattern of lamellipodial wrinkles. In support of the numerical modeling findings, we observed that the transient halting of retraction at the rear using micromanipulation induced the formation of lamellipodial wrinkles in previously flat lamellipodia. The obtained results suggest that the conspicuous lamellipodial wrinkles observed in migrating fish keratocytes are the product of transient imbalances between front and rear displacements, and are therefore useful markers of the short scale dynamics of extension and retraction coordination during cell migration.
Collapse
|
44
|
Simoncelli S, Makarova M, Wardley W, Owen DM. Toward an Axial Nanoscale Ruler for Fluorescence Microscopy. ACS NANO 2017; 11:11762-11767. [PMID: 29161014 DOI: 10.1021/acsnano.7b07133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In the discussion of resolution in optical microscopy, axial precision has often come second to its lateral counterpart. However, biological systems make no special arrangements for our preferred direction of imaging. The ability to measure axial distances, that is, the heights of fluorophores relative to a plane of reference, is thus of paramount importance and has been the subject of several recent advances. A novel method is to modify the fluorescence emission based on the height of the individual fluorophore, such that its z-position is encoded somehow in the detected signal. One such approach is metal-enhanced energy transfer, recently extended to multicolor distance measurements and applied to study the topography of the nuclear membrane. Here, the fluorescence lifetime is shortened due to the proximity of the fluorophores to a thin metallic surface. Fluorescence lifetime imaging can therefore be used as an axial ruler with nanometer precision.
Collapse
Affiliation(s)
- Sabrina Simoncelli
- Blackett Laboratory, Department of Physics, Imperial College London , London SW7 2AZ, United Kingdom
| | - Maria Makarova
- Francis Crick Institute and Randall Division of Cell and Molecular Biophysics, King's College London , London NW1 1AT, United Kingdom
| | - William Wardley
- Department of Physics, King's College London , London WC2R 2LS, United Kingdom
| | - Dylan M Owen
- Department of Physics and Randall Division of Cell and Molecular Biophysics, King's College London , London SE1 1UL, United Kingdom
| |
Collapse
|
45
|
Gaertner F, Ahmad Z, Rosenberger G, Fan S, Nicolai L, Busch B, Yavuz G, Luckner M, Ishikawa-Ankerhold H, Hennel R, Benechet A, Lorenz M, Chandraratne S, Schubert I, Helmer S, Striednig B, Stark K, Janko M, Böttcher RT, Verschoor A, Leon C, Gachet C, Gudermann T, Mederos Y Schnitzler M, Pincus Z, Iannacone M, Haas R, Wanner G, Lauber K, Sixt M, Massberg S. Migrating Platelets Are Mechano-scavengers that Collect and Bundle Bacteria. Cell 2017; 171:1368-1382.e23. [PMID: 29195076 DOI: 10.1016/j.cell.2017.11.001] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/27/2017] [Accepted: 10/30/2017] [Indexed: 01/07/2023]
Abstract
Blood platelets are critical for hemostasis and thrombosis and play diverse roles during immune responses. Despite these versatile tasks in mammalian biology, their skills on a cellular level are deemed limited, mainly consisting in rolling, adhesion, and aggregate formation. Here, we identify an unappreciated asset of platelets and show that adherent platelets use adhesion receptors to mechanically probe the adhesive substrate in their local microenvironment. When actomyosin-dependent traction forces overcome substrate resistance, platelets migrate and pile up the adhesive substrate together with any bound particulate material. They use this ability to act as cellular scavengers, scanning the vascular surface for potential invaders and collecting deposited bacteria. Microbe collection by migrating platelets boosts the activity of professional phagocytes, exacerbating inflammatory tissue injury in sepsis. This assigns platelets a central role in innate immune responses and identifies them as potential targets to dampen inflammatory tissue damage in clinical scenarios of severe systemic infection.
Collapse
Affiliation(s)
- Florian Gaertner
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), 13347 Berlin, Germany.
| | - Zerkah Ahmad
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Gerhild Rosenberger
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Shuxia Fan
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Benjamin Busch
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU, 80336 Munich, Germany
| | - Gökce Yavuz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Manja Luckner
- Ultrastructural Research, Department Biology I, Biozentrum, Ludwig-Maximillians-Universität, 82152 Martinsried, Germany
| | | | - Roman Hennel
- Department of Radiation Oncology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Alexandre Benechet
- Division of Immunology, Transplantation and Infectious Diseases, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Sue Chandraratne
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Irene Schubert
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Sebastian Helmer
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Bianca Striednig
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), 13347 Berlin, Germany
| | - Marek Janko
- Department of Materials Science, Technische Universität, 64287 Darmstadt, Germany
| | - Ralph T Böttcher
- Max-Planck Institute of Biochemistry, 82152 Martinsried, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), 13347 Berlin, Germany
| | - Admar Verschoor
- Institute for Systemic Inflammation Research, Universität zu Lübeck, 23538 Lübeck, Germany
| | - Catherine Leon
- UMR S949, Inserm, Université de Strasbourgh, Etablissement Français du Sang-Alsace, 67065 Strasbourg, France
| | - Christian Gachet
- UMR S949, Inserm, Université de Strasbourgh, Etablissement Français du Sang-Alsace, 67065 Strasbourg, France
| | - Thomas Gudermann
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität, 80336 Munich, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), 13347 Berlin, Germany
| | - Michael Mederos Y Schnitzler
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität, 80336 Munich, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), 13347 Berlin, Germany
| | - Zachary Pincus
- Department of Developmental Biology and Department of Genetics, Washington University, St. Louis, St. Louis, MO 63110, USA
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Rainer Haas
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU, 80336 Munich, Germany; German Center for Infection Research (DZIF), Munich Site, 80336 Munich, Germany
| | - Gerhard Wanner
- Ultrastructural Research, Department Biology I, Biozentrum, Ludwig-Maximillians-Universität, 82152 Martinsried, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Michael Sixt
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, 81377 Munich, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), 13347 Berlin, Germany.
| |
Collapse
|
46
|
Ferrera P, Zepeda A, Arias C. Nonsteroidal anti-inflammatory drugs attenuate amyloid-β protein-induced actin cytoskeletal reorganization through Rho signaling modulation. Cell Mol Neurobiol 2017; 37:1311-1318. [PMID: 28124209 PMCID: PMC11482099 DOI: 10.1007/s10571-017-0467-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/19/2017] [Indexed: 01/15/2023]
Abstract
Amyloid-β protein (Aβ) neurotoxicity occurs along with the reorganization of the actin-cytoskeleton through the activation of the Rho GTPase pathway. In addition to the classical mode of action of the non-steroidal anti-inflammatory drugs (NSAIDs), indomethacin, and ibuprofen have Rho-inhibiting effects. In order to evaluate the role of the Rho GTPase pathway on Aβ-induced neuronal death and on neuronal morphological modifications in the actin cytoskeleton, we explored the role of NSAIDS in human-differentiated neuroblastoma cells exposed to Aβ. We found that Aβ induced neurite retraction and promoted the formation of different actin-dependent structures such as stress fibers, filopodia, lamellipodia, and ruffles. In the presence of Aβ, both NSAIDs prevented neurite collapse and formation of stress fibers without affecting the formation of filopodia and lamellipodia. Similar results were obtained when the downstream effector, Rho kinase inhibitor Y27632, was applied in the presence of Aβ. These results demonstrate the potential benefits of the Rho-inhibiting NSAIDs in reducing Aβ-induced effects on neuronal structural alterations.
Collapse
Affiliation(s)
- Patricia Ferrera
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico.
| |
Collapse
|
47
|
Chang CH, Lee HH, Lee CH. Substrate properties modulate cell membrane roughness by way of actin filaments. Sci Rep 2017; 7:9068. [PMID: 28831175 PMCID: PMC5567215 DOI: 10.1038/s41598-017-09618-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 07/24/2017] [Indexed: 01/09/2023] Open
Abstract
Cell membrane roughness has been proposed as a sensitive feature to reflect cellular physiological conditions. In order to know whether membrane roughness is associated with the substrate properties, we employed the non-interferometric wide-field optical profilometry (NIWOP) technique to measure the membrane roughness of living mouse embryonic fibroblasts with different conditions of the culture substrate. By controlling the surface density of fibronectin (FN) coated on the substrate, we found that cells exhibited higher membrane roughness as the FN density increased in company with larger focal adhesion (FA) sizes. The examination of membrane roughness was also confirmed with atomic force microscopy. Using reagents altering actin or microtubule cytoskeletons, we provided evidence that the dynamics of actin filaments rather than that of microtubules plays a crucial role for the regulation of membrane roughness. By changing the substrate rigidity, we further demonstrated that the cells seeded on compliant gels exhibited significantly lower membrane roughness and smaller FAs than the cells on rigid substrate. Taken together, our data suggest that the magnitude of membrane roughness is modulated by way of actin dynamics in cells responding to substrate properties.
Collapse
Affiliation(s)
- Chao-Hung Chang
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Hsiao-Hui Lee
- Department of Life Sciences & Institute of Genome Sciences, National Yang-Ming University, Taipei, 11221, Taiwan.
| | - Chau-Hwang Lee
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan. .,Institute of Biophotonics, National Yang-Ming University, Taipei, 11221, Taiwan. .,Department of Physics, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
48
|
Gallagher-Jones M, Dias CSB, Pryor A, Bouchmella K, Zhao L, Lo YH, Cardoso MB, Shapiro D, Rodriguez J, Miao J. Correlative cellular ptychography with functionalized nanoparticles at the Fe L-edge. Sci Rep 2017; 7:4757. [PMID: 28684732 PMCID: PMC5500580 DOI: 10.1038/s41598-017-04784-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/19/2017] [Indexed: 12/17/2022] Open
Abstract
Precise localization of nanoparticles within a cell is crucial to the understanding of cell-particle interactions and has broad applications in nanomedicine. Here, we report a proof-of-principle experiment for imaging individual functionalized nanoparticles within a mammalian cell by correlative microscopy. Using a chemically-fixed HeLa cell labeled with fluorescent core-shell nanoparticles as a model system, we implemented a graphene-oxide layer as a substrate to significantly reduce background scattering. We identified cellular features of interest by fluorescence microscopy, followed by scanning transmission X-ray tomography to localize the particles in 3D, and ptychographic coherent diffractive imaging of the fine features in the region at high resolution. By tuning the X-ray energy to the Fe L-edge, we demonstrated sensitive detection of nanoparticles composed of a 22 nm magnetic Fe3O4 core encased by a 25-nm-thick fluorescent silica (SiO2) shell. These fluorescent core-shell nanoparticles act as landmarks and offer clarity in a cellular context. Our correlative microscopy results confirmed a subset of particles to be fully internalized, and high-contrast ptychographic images showed two oxidation states of individual nanoparticles with a resolution of ~16.5 nm. The ability to precisely localize individual fluorescent nanoparticles within mammalian cells will expand our understanding of the structure/function relationships for functionalized nanoparticles.
Collapse
Affiliation(s)
- Marcus Gallagher-Jones
- Department of Physics and Astronomy and California NanoSystems Institute, University of California Los Angeles, California, 90095, USA
| | - Carlos Sato Baraldi Dias
- Department of Physics and Astronomy and California NanoSystems Institute, University of California Los Angeles, California, 90095, USA
| | - Alan Pryor
- Department of Physics and Astronomy and California NanoSystems Institute, University of California Los Angeles, California, 90095, USA
| | - Karim Bouchmella
- Brazilian Synchrotron Laboratory (LNLS), Brazilian Center for in Energy and Materials, CEP 13083-970, Campinas, São Paulo, Brazil
| | - Lingrong Zhao
- Department of Physics and Astronomy and California NanoSystems Institute, University of California Los Angeles, California, 90095, USA
- Department of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuan Hung Lo
- Department of Physics and Astronomy and California NanoSystems Institute, University of California Los Angeles, California, 90095, USA
| | - Mateus Borba Cardoso
- Brazilian Synchrotron Laboratory (LNLS), Brazilian Center for in Energy and Materials, CEP 13083-970, Campinas, São Paulo, Brazil
| | - David Shapiro
- Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, California, 94720, USA
| | - Jose Rodriguez
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, California, 90095, USA.
| | - Jianwei Miao
- Department of Physics and Astronomy and California NanoSystems Institute, University of California Los Angeles, California, 90095, USA.
| |
Collapse
|
49
|
Martin KC, Yuan X, Stimac G, Bannerman K, Anderson J, Roy C, Glykofrydis F, Yin H, Davies JA. Symmetry-breaking in branching epithelia: cells on micro-patterns under flow challenge the hypothesis of positive feedback by a secreted autocrine inhibitor of motility. J Anat 2017; 230:766-774. [PMID: 28369863 PMCID: PMC5442143 DOI: 10.1111/joa.12599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2017] [Indexed: 01/21/2023] Open
Abstract
Branching morphogenesis of epithelia involves division of cells into leader (tip) and follower (stalk) cells. Published work on cell lines in culture has suggested that symmetry-breaking takes place via a secreted autocrine inhibitor of motility, the inhibitor accumulating more in concave regions of the culture boundary, slowing advance of cells there, and less in convex areas, allowing advance and a further exaggeration of the concave/convex difference. Here we test this hypothesis using a two-dimensional culture system that includes strong flow conditions to remove accumulating diffusible secretions. We find that, while motility does indeed follow boundary curvature in this system, flow makes no difference: this challenges the hypothesis of control by a diffusible secreted autocrine inhibitor.
Collapse
Affiliation(s)
- Kimberly C. Martin
- Centre for Integrative PhysiologyUniversity of EdinburghGeorge SquareEdinburghEH8 9XBUK
| | - Xiaofei Yuan
- School of EngineeringJames Watt BuildingUniversity of GlasgowGL12 8QQUK
| | - Gregory Stimac
- Centre for Integrative PhysiologyUniversity of EdinburghGeorge SquareEdinburghEH8 9XBUK
| | - Kieran Bannerman
- Centre for Integrative PhysiologyUniversity of EdinburghGeorge SquareEdinburghEH8 9XBUK
| | - Jamie Anderson
- Centre for Integrative PhysiologyUniversity of EdinburghGeorge SquareEdinburghEH8 9XBUK
| | - Chloe Roy
- Centre for Integrative PhysiologyUniversity of EdinburghGeorge SquareEdinburghEH8 9XBUK
| | - Fokion Glykofrydis
- Centre for Integrative PhysiologyUniversity of EdinburghGeorge SquareEdinburghEH8 9XBUK
| | - Huabing Yin
- School of EngineeringJames Watt BuildingUniversity of GlasgowGL12 8QQUK
| | - Jamie A. Davies
- Centre for Integrative PhysiologyUniversity of EdinburghGeorge SquareEdinburghEH8 9XBUK
| |
Collapse
|
50
|
Salameh TS, Shah GN, Price TO, Hayden MR, Banks WA. Blood-Brain Barrier Disruption and Neurovascular Unit Dysfunction in Diabetic Mice: Protection with the Mitochondrial Carbonic Anhydrase Inhibitor Topiramate. J Pharmacol Exp Ther 2016; 359:452-459. [PMID: 27729477 PMCID: PMC5118649 DOI: 10.1124/jpet.116.237057] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/23/2016] [Indexed: 12/19/2022] Open
Abstract
All forms of diabetes mellitus are characterized by chronic hyperglycemia, resulting in the development of a number of microvascular and macrovascular pathologies. Diabetes is also associated with changes in brain microvasculature, leading to dysfunction and ultimately disruption of the blood-brain barrier (BBB). These changes are correlated with a decline in cognitive function. In diabetes, BBB damage is associated with increased oxidative stress and reactive oxygen species. This occurs because of the increased oxidative metabolism of glucose caused by hyperglycemia. Decreasing the production of bicarbonate with the use of a mitochondrial carbonic anhydrase inhibitor (mCAi) limits oxidative metabolism and the production of reactive oxygen species. In this study, we have demonstrated that 1) streptozotocin-induced diabetes resulted in BBB disruption, 2) ultrastructural studies showed a breakdown of the BBB and changes to the neurovascular unit (NVU), including a loss of brain pericytes and retraction of astrocytes, the two cell types that maintain the BBB, and 3) treatment with topiramate, a mCAi, attenuated the effects of diabetes on BBB disruption and ultrastructural changes in the neurovascular unit.
Collapse
Affiliation(s)
- Therese S Salameh
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington (T.S.S., W.A.B.); Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington (T.S.S., W.A.B.); Division of Endocrinology, Department of Internal Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri (G.N.S., T.O.P.); Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Missouri, Columbia, Missouri (M.R.H.); Diabetes and Cardiovascular Research Laboratory, University of Missouri, Columbia, Missouri (M.H.R.)
| | - Gul N Shah
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington (T.S.S., W.A.B.); Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington (T.S.S., W.A.B.); Division of Endocrinology, Department of Internal Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri (G.N.S., T.O.P.); Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Missouri, Columbia, Missouri (M.R.H.); Diabetes and Cardiovascular Research Laboratory, University of Missouri, Columbia, Missouri (M.H.R.)
| | - Tulin O Price
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington (T.S.S., W.A.B.); Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington (T.S.S., W.A.B.); Division of Endocrinology, Department of Internal Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri (G.N.S., T.O.P.); Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Missouri, Columbia, Missouri (M.R.H.); Diabetes and Cardiovascular Research Laboratory, University of Missouri, Columbia, Missouri (M.H.R.)
| | - Melvin R Hayden
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington (T.S.S., W.A.B.); Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington (T.S.S., W.A.B.); Division of Endocrinology, Department of Internal Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri (G.N.S., T.O.P.); Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Missouri, Columbia, Missouri (M.R.H.); Diabetes and Cardiovascular Research Laboratory, University of Missouri, Columbia, Missouri (M.H.R.)
| | - William A Banks
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington (T.S.S., W.A.B.); Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington (T.S.S., W.A.B.); Division of Endocrinology, Department of Internal Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri (G.N.S., T.O.P.); Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Missouri, Columbia, Missouri (M.R.H.); Diabetes and Cardiovascular Research Laboratory, University of Missouri, Columbia, Missouri (M.H.R.)
| |
Collapse
|