1
|
Sharip A, Kunz J. Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis Progression and Therapy. Cells 2025; 14:266. [PMID: 39996739 PMCID: PMC11854242 DOI: 10.3390/cells14040266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Liver fibrosis, a consequence of chronic liver injury, represents a major global health burden and is the leading cause of liver failure, morbidity, and mortality. The pathological hallmark of this condition is excessive extracellular matrix deposition, driven primarily by integrin-mediated mechanotransduction. Integrins, transmembrane heterodimeric proteins that serve as primary ECM receptors, orchestrate complex mechanosignaling networks that regulate the activation, differentiation, and proliferation of hepatic stellate cells and other ECM-secreting myofibroblasts. These mechanical signals create self-reinforcing feedback loops that perpetuate the fibrotic response. Recent advances have provided insight into the roles of specific integrin subtypes in liver fibrosis and revealed their regulation of key downstream effectors-including transforming growth factor beta, focal adhesion kinase, RhoA/Rho-associated, coiled-coil containing protein kinase, and the mechanosensitive Hippo pathway. Understanding these mechanotransduction networks has opened new therapeutic possibilities through pharmacological manipulation of integrin-dependent signaling.
Collapse
Affiliation(s)
- Aigul Sharip
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
- Laboratory of Bioinformatics and Systems Biology, National Laboratory Astana, Astana 020000, Kazakhstan
| | - Jeannette Kunz
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
| |
Collapse
|
2
|
Dutta Gupta S, Pal N, Ta M. Vitronectin regulates focal adhesion turnover and migration of human placenta-derived MSCs under nutrient stress. Eur J Cell Biol 2025; 104:151477. [PMID: 39893799 DOI: 10.1016/j.ejcb.2025.151477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
At sites of tissue damage and wound healing, the mesenchymal stem cells (MSCs) are often challenged by nutrient availability due to blood supply disruption. Thus, it becomes critical to identify novel factors and their mechanism of action in regulating the adhesion and migration of MSCs under nutrient stress condition for successful clinical application. In human placenta-derived MSCs (PL-MSCs), we demonstrated an increase in cell spread area, along with increased adhesion and reduced migration of the cells, when cultured under nutrient stress condition. Correspondingly, an increase in the total number per cell and size of focal adhesions (FAs), together with prominent stress fibers were observed in nutrient-stressed PL-MSCs compared to control PL-MSCs. The FAs were demonstrated to be more stable, exhibiting slower turnover and longer lifespan. Vitronectin (VTN), an ECM glycoprotein, was upregulated under nutrient stress condition. Knockdown of VTN in PL-MSCs led to a significant reduction in the total number per cell and size of FAs, along with their faster turnover and shorter lifespan. Subsequently, a reversal in the cell spread area, adhesion and migration properties of the nutrient-stressed PL-MSCs were noted. Additionally, our findings indicated that VTN, as an upstream regulator, stimulated the phosphorylation of myosin light chain, which possibly promoted the maturation and stability of FAs along with assembly of stress fibers, thereby leading to increased adhesion and reduced migration of the cells. Overall, our study defines a distinct role of VTN as a critical regulator of migration in PL-MSCs under nutrient stress condition.
Collapse
Affiliation(s)
- Srishti Dutta Gupta
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| | - Nitish Pal
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| | - Malancha Ta
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| |
Collapse
|
3
|
Meinung CP, Boi L, Pandamooz S, Mazaud D, Ghézali G, Rouach N, Neumann ID. OXTR-mediated signaling in astrocytes contributes to anxiolysis. Mol Psychiatry 2024:10.1038/s41380-024-02870-5. [PMID: 39702695 DOI: 10.1038/s41380-024-02870-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/13/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Astrocytes are an indispensable part of signal processing within the mammalian brain. Thus, the mode of action of a neuropeptide such as oxytocin (OXT) can only be fully understood considering this integral part of the CNS. Here, we show that OXT regulates astrocytic gene expression, intracellular signaling and specific proteins both in vitro and in vivo. This translates into rapid regulation of astroglial structural and functional properties including cytoskeletal plasticity, coverage of synapses and gap-junction coupling. At the molecular level, we identify the previously undescribed Sp1-Gem signaling cascade as the key driver for these cell type-specific OXT effects. Finally at the behavioral level, we found in vivo that OXT requires astrocytes to exert its well described anxiolytic properties within the hypothalamic paraventricular nucleus. Thus, our study points to OXT receptor-expressing astrocytes as a critical component of the brain OXT system.
Collapse
Affiliation(s)
- Carl-Philipp Meinung
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Laura Boi
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Sareh Pandamooz
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - David Mazaud
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Grégory Ghézali
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
Matsumura F, Murayama T, Kuriyama R, Matsumura A, Yamashiro S. Myosin phosphatase targeting subunit1 controls localization and motility of Rab7-containing vesicles: Is myosin phosphatase a cytoplasmic dynein regulator? Cytoskeleton (Hoboken) 2024; 81:872-882. [PMID: 38700016 PMCID: PMC11615836 DOI: 10.1002/cm.21871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024]
Abstract
Myosin phosphatase targeting subunit1 (MYPT1) is a critical subunit of myosin phosphatase (MP), which brings PP1Cδ phosphatase and its substrate together. We previously showed that MYPT1 depletion resulted in oblique chromatid segregation. Therefore, we hypothesized that MYPT1 may control microtubule-dependent motor activity. Dynein, a minus-end microtubule motor, is known to be involved in mitotic spindle assembly. We thus examined whether MYPT1 and dynein may interact. Proximity ligation assay and co-immunoprecipitation revealed that MYPT1 and dynein intermediate chain (DIC) were associated. We found that DIC phosphorylation is increased in MYPT1-depleted cells in vivo, and that MP was able to dephosphorylate DIC in vitro. MYPT1 depletion also altered the localization and motility of Rab7-containing vesicles. MYPT1-depletion dispersed the perinuclear Rab7 localization to the peripheral in interphase cells. The dispersed Rab7 localization was rescued by microinjection of a constitutively active, truncated MYPT1 mutant, supporting that MP is responsible for the altered Rab7 localization. Analyses of Rab7 vesicle trafficking also revealed that minus-end transport was reduced in MYPT1-depleted cells. These results suggest an unexpected role of MP: MP controls dynein activity in both mitotic and interphase cells, possibly by dephosphorylating dynein subunits including DIC.
Collapse
Affiliation(s)
- Fumio Matsumura
- Department of Molecular Biology & BiochemistryRutgers UniversityPiscatawayNew JerseyUSA
| | - Takashi Murayama
- Department of PharmacologyJuntendo University School of MedicineTokyoJapan
| | - Ryoko Kuriyama
- Department of Genetics, Cell Biology and DevelopmentUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Aya Matsumura
- Department of Molecular Biology & BiochemistryRutgers UniversityPiscatawayNew JerseyUSA
| | - Shigeko Yamashiro
- Department of Molecular Biology & BiochemistryRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
5
|
Toran P, Novelli A, Lazor J, Vachon A, Wojchowski DM. Regulation of BCR-dependent germinal center B-cell formation by HGAL and insight into its emerging myeloid ortholog, C1ORF150. Front Immunol 2024; 15:1437516. [PMID: 39474423 PMCID: PMC11518702 DOI: 10.3389/fimmu.2024.1437516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/24/2024] [Indexed: 11/15/2024] Open
Abstract
The specificity of cytokine and immunoreceptor signaling frequently depends upon receptor recruitment of select adaptor proteins and specifically engaged effectors. This review focuses on the orthologous adaptor proteins, HGAL and C1ORF150, and aims to provide insight into their respective modulation of lymphoid and myeloid cell signaling, formation, and function. HGAL acts predominantly within germinal center B cells as an important BCR signal transducer. Effects on BCR signalosome assembly involve HGAL's localization to the plasma membrane via its lipidation, initial interactions with SYK, the pY-phosphorylation of HGAL including its recruitment of GRB2, and HGAL engagement of PDZ-RhoGEF and RhoA signaling. At ligated BCRs, this includes HGAL(-GRB2) stimulation of SYK kinase, attenuation of calcium flux-dependent and NF-κB expression, promotion of cSMAC formation, and cytoskeletal remodeling associated with HGAL-attenuated cell migration. HGAL and partnered effectors also impact on DLBCL pathogenesis, and studies are summarized on HGAL's actions (using DLBCL and Burkitt lymphoma B cells) including cell migration effects, HGAL modulation of cytoskeletal components, and insightful HGAL transgenic mouse and xenograft models. For C1ORF150, its HGAL-homologous subdomains are considered, together with studies that demonstrate C1OR150's FcϵRI- and KIT-mediated expression and phosphorylation in primary human mast cells. Intriguingly, recent GWAS studies have identified a C1ORF150 in-frame splice variant that is strongly associated with urticaria. Candidate mechanisms via which the encoded "C1ORF150-Δexon2" isoform affects mast cell degranulation are considered, including FcϵR1 and/or KIT receptor connections, and candidate "myristoylation switch" mechanisms.
Collapse
Affiliation(s)
- Paul Toran
- Department of Molecular, Cellular and Biomedical Sciences University of New Hampshire, Durham, NH, United States
| | - Anthony Novelli
- Department of Molecular, Cellular and Biomedical Sciences University of New Hampshire, Durham, NH, United States
| | - Jennifer Lazor
- Department of Molecular, Cellular and Biomedical Sciences University of New Hampshire, Durham, NH, United States
- Biotherapeutics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, United States
| | - Alexandra Vachon
- Department of Molecular, Cellular and Biomedical Sciences University of New Hampshire, Durham, NH, United States
| | - Don M. Wojchowski
- Department of Molecular, Cellular and Biomedical Sciences University of New Hampshire, Durham, NH, United States
| |
Collapse
|
6
|
Weißenbruch K, Mayor R. Actomyosin forces in cell migration: Moving beyond cell body retraction. Bioessays 2024; 46:e2400055. [PMID: 39093597 DOI: 10.1002/bies.202400055] [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: 03/12/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
In textbook illustrations of migrating cells, actomyosin contractility is typically depicted as the contraction force necessary for cell body retraction. This dogma has been transformed by the molecular clutch model, which acknowledges that actomyosin traction forces also generate and transmit biomechanical signals at the leading edge, enabling cells to sense and shape their migratory path in mechanically complex environments. To fulfill these complementary functions, the actomyosin system assembles a gradient of contractile energy along the front-rear axis of migratory cells. Here, we highlight the hierarchic assembly and self-regulatory network structure of the actomyosin system and explain how the kinetics of different nonmuscle myosin II (NM II) paralogs synergize during contractile force generation. Our aim is to emphasize how protrusion formation, cell adhesion, contraction, and retraction are spatiotemporally integrated during different modes of migration, including chemotaxis and durotaxis. Finally, we hypothesize how different NM II paralogs might tune aspects of migration in vivo, highlighting future research directions.
Collapse
Affiliation(s)
- Kai Weißenbruch
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
7
|
Marshall-Burghardt S, Migueles-Ramírez RA, Lin Q, El Baba N, Saada R, Umar M, Mavalwala K, Hayer A. Excitable Rho dynamics control cell shape and motility by sequentially activating ERM proteins and actomyosin contractility. SCIENCE ADVANCES 2024; 10:eadn6858. [PMID: 39241071 PMCID: PMC11378911 DOI: 10.1126/sciadv.adn6858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/31/2024] [Indexed: 09/08/2024]
Abstract
Migration of endothelial and many other cells requires spatiotemporal regulation of protrusive and contractile cytoskeletal rearrangements that drive local cell shape changes. Unexpectedly, the small GTPase Rho, a crucial regulator of cell movement, has been reported to be active in both local cell protrusions and retractions, raising the question of how Rho activity can coordinate cell migration. Here, we show that Rho activity is absent in local protrusions and active during retractions. During retractions, Rho rapidly activated ezrin-radixin-moesin proteins (ERMs) to increase actin-membrane attachment, and, with a delay, nonmuscle myosin 2 (NM2). Rho activity was excitable, with NM2 acting as a slow negative feedback regulator. Strikingly, inhibition of SLK/LOK kinases, through which Rho activates ERMs, caused elongated cell morphologies, impaired Rho-induced cell contractions, and reverted Rho-induced blebbing. Together, our study demonstrates that Rho activity drives retractions by sequentially enhancing ERM-mediated actin-membrane attachment for force transmission and NM2-dependent contractility.
Collapse
Affiliation(s)
- Seph Marshall-Burghardt
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- Graduate Program in Biology, McGill University, Montréal, Québec, Canada
| | - Rodrigo A Migueles-Ramírez
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- PhD Program in Quantitative Life Sciences, McGill University, Montréal, Québec, Canada
| | - Qiyao Lin
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- Graduate Program in Biology, McGill University, Montréal, Québec, Canada
| | - Nada El Baba
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- Graduate Program in Biology, McGill University, Montréal, Québec, Canada
| | - Rayan Saada
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| | - Mustakim Umar
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| | - Kian Mavalwala
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| | - Arnold Hayer
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| |
Collapse
|
8
|
Del Caño LR, South AP, O'Toole EA, Kelsell DP, Blaydon DC. A Role for Aquaporin-5 Variants in Regulation of the Actin Cytoskeleton in Non-Epidermolytic Palmoplantar Keratoderma. J Invest Dermatol 2024; 144:2092-2096. [PMID: 38527693 DOI: 10.1016/j.jid.2024.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/27/2024]
Affiliation(s)
- Laura Ramos Del Caño
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, The Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Andrew P South
- Department of Dermatology & Cutaneous Biology, Thomas Jefferson University, 233 South Tenth Street BLSB 406, Philadelphia, Pennsylvania, USA
| | - Edel A O'Toole
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, The Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - David P Kelsell
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, The Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Diana C Blaydon
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, The Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
9
|
Hossen F, Sun GY, Lee JC. Oligomeric Tau-induced oxidative damage and functional alterations in cerebral endothelial cells: Role of RhoA/ROCK signaling pathway. Free Radic Biol Med 2024; 221:261-272. [PMID: 38815773 PMCID: PMC11184584 DOI: 10.1016/j.freeradbiomed.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Despite of yet unknown mechanism, microvascular deposition of oligomeric Tau (oTau) has been implicated in alteration of the Blood-Brain Barrier (BBB) function in Alzheimer's disease (AD) brains. In this study, we employed an in vitro BBB model using primary mouse cerebral endothelial cells (CECs) to investigate the mechanism underlying the effects of oTau on BBB function. We found that exposing CECs to oTau induced oxidative stress through NADPH oxidase, increased oxidative damage to proteins, decreased proteasome activity, and expressions of tight junction (TJ) proteins including occludin, zonula occludens-1 (ZO-1) and claudin-5. These effects were suppressed by the pretreatment with Fasudil, a RhoA/ROCK signaling inhibitor. Consistent with the biochemical alterations, we found that exposing the basolateral side of CECs to oTau in the BBB model disrupted the integrity of the BBB, as indicated by an increase in FITC-dextran transport across the model, and a decrease in trans endothelial electrical resistance (TEER). oTau also increased the transmigration of peripheral blood mononuclear cells (PBMCs) in the BBB model. These functional alterations in the BBB induced by oTau were also suppressed by Fasudil. Taken together, our findings suggest that targeting the RhoA/ROCK pathway can be a potential therapeutic strategy to maintain BBB function in AD.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, 65211, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
10
|
Tsuji-Tamura K, Sato M, Tamura M. Pharmacological control of angiogenesis by regulating phosphorylation of myosin light chain 2. Cell Signal 2024; 120:111223. [PMID: 38729320 DOI: 10.1016/j.cellsig.2024.111223] [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: 11/15/2023] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Control of angiogenesis is widely considered a therapeutic strategy, but reliable control methods are still under development. Phosphorylation of myosin light chain 2 (MLC2), which regulates actin-myosin interaction, is critical to the behavior of vascular endothelial cells (ECs) during angiogenesis. MLC2 is phosphorylated by MLC kinase (MLCK) and dephosphorylated by MLC phosphatase (MLCP) containing a catalytic subunit PP1. We investigated the potential role of MLC2 in the pharmacological control of angiogenesis. METHODS AND RESULTS We exposed transgenic zebrafish Tg(fli1a:Myr-mCherry)ncv1 embryos to chemical inhibitors and observed vascular development. PP1 inhibition by tautomycetin increased length of intersegmental vessels (ISVs), whereas MLCK inhibition by ML7 decreased it; these effects were not accompanied by structural dysplasia. ROCK inhibition by Y-27632 also decreased vessel length. An in vitro angiogenesis model of human umbilical vein endothelial cells (HUVECs) showed that tautomycetin increased vascular cord formation, whereas ML7 and Y-27632 decreased it. These effects appear to be influenced by regulation of cell morphology rather than cell viability or motility. Actin co-localized with phosphorylated MLC2 (pMLC2) was abundant in vascular-like elongated-shaped ECs, but poor in non-elongated ECs. pMLC2 was associated with tightly arranged actin, but not with loosely arranged actin. Moreover, knockdown of MYL9 gene encoding MLC2 reduced total MLC2 and pMLC2 protein and inhibited angiogenesis in HUVECs. CONCLUSION The present study found that MLC2 is a pivotal regulator of angiogenesis. MLC2 phosphorylation may be involved in the regulation of of cell morphogenesis and cell elongation. The functionally opposite inhibitors positively or negatively control angiogenesis, probably through the regulating EC morphology. These findings may provide a unique therapeutic target for angiogenesis.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan.
| | - Mari Sato
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan
| | - Masato Tamura
- Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13, Nishi 7, Kita-Ku, Sapporo 060-8586, Japan
| |
Collapse
|
11
|
Peng T, Yang S, Lian W, Liu X, Zheng P, Qin X, Liao B, Zhou P, Wang Y, Liu F, Yang Z, Ye Z, Shan H, Liu X, Yu Y, Li R. Cytoskeletal and inter-cellular junction remodelling in endometrial organoids under oxygen-glucose deprivation: a new potential pathological mechanism for thin endometria. Hum Reprod 2024; 39:1778-1793. [PMID: 38915267 DOI: 10.1093/humrep/deae137] [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: 01/03/2024] [Revised: 05/21/2024] [Indexed: 06/26/2024] Open
Abstract
STUDY QUESTION What is the pathological mechanism involved in a thin endometrium, particularly under ischaemic conditions? SUMMARY ANSWER Endometrial dysfunction in patients with thin endometrium primarily results from remodelling in cytoskeletons and cellular junctions of endometrial epithelial cells under ischemic conditions. WHAT IS KNOWN ALREADY A healthy endometrium is essential for successful embryo implantation and subsequent pregnancy; ischemic conditions in a thin endometrium compromise fertility outcomes. STUDY DESIGN, SIZE, DURATION We recruited 10 patients with thin endometrium and 15 patients with healthy endometrium. Doppler ultrasound and immunohistochemical results confirmed the presence of insufficient endometrial blood perfusion in patients with thin endometrium. Organoids were constructed using healthy endometrial tissue and cultured under oxygen-glucose deprivation (OGD) conditions for 24 h. The morphological, transcriptomic, protein expression, and signaling pathway changes in the OGD organoids were observed. These findings were validated in both thin endometrial tissue and healthy endometrial tissue samples. PARTICIPANTS/MATERIALS, SETTING, METHODS Endometrial thickness and blood flow were measured during the late follicular phase using transvaginal Doppler ultrasound. Endometrial tissue was obtained via hysteroscopy. Fresh endometrial tissues were used for the generation and culture of human endometrial organoids. Organoids were cultured in an appropriate medium and subjected to OGD to simulate ischemic conditions. Apoptosis and cell death were assessed using Annexin-V/propidium iodide staining. Immunofluorescence analysis, RNA sequencing, western blotting, simple westerns, immunohistochemistry, and electron microscopy were conducted to evaluate cellular and molecular changes. MAIN RESULTS AND THE ROLE OF CHANCE Patients with thin endometrium showed significantly reduced endometrial thickness and altered blood flow patterns compared to those with healthy endometrium. Immunohistochemical staining revealed fewer CD34-positive blood vessels and glands in the thin endometrium group. Organoids cultured under OGD conditions exhibited significant morphological changes, increased apoptosis, and cell death. RNA-seq identified differentially expressed genes related to cytoskeletal remodeling and stress responses. OGD induced a strong cytoskeletal reorganization, mediated by the RhoA/ROCK signaling pathway. Additionally, electron microscopy indicated compromised epithelial integrity and abnormal cell junctions in thin endometrial tissues. Upregulation of hypoxia markers (HIF-1α and HIF-2α) and activation of the RhoA/ROCK pathway were also observed in thin endometrial tissues, suggesting ischemia and hypoxia as underlying mechanisms. LARGE SCALE DATA none. LIMITATIONS AND REASONS FOR CAUTION The study was conducted in an in vitro model, which may not fully replicate the complexity of in vivo conditions. WIDER IMPLICATIONS OF THE FINDINGS This research provides a new three-dimensional in vitro model of thin endometrium, as well as novel insights into the pathophysiological mechanisms of endometrial ischaemia in thin endometrium, offering potential avenues for identifying therapeutic targets for treating fertility issues related to thin endometrium. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the National Natural Science Foundation of China (81925013); National Key Research and Development Project of China (2022YFC2702500, 2021YFC2700303, 2021YFC2700601); the Capital Health Research and Development Project (SF2022-1-4092); the National Natural Science Foundation of China (82288102, 81925013, 82225019, 82192873); Special Project on Capital Clinical Diagnosis and Treatment Technology Research and Transformation Application (Z211100002921054); the Frontiers Medical Center, Tianfu Jincheng Laboratory Foundation(TFJC2023010001). The authors declare that no competing interests exist.
Collapse
Affiliation(s)
- TianLiu Peng
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Shuo Yang
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Weisi Lian
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Xiaojuan Liu
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Ping Zheng
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Xunsi Qin
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Baoying Liao
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Ping Zhou
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Yue Wang
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Fenting Liu
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Zi Yang
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Zhenhong Ye
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Hongying Shan
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Xiyao Liu
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| | - Yang Yu
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Rong Li
- Department of Obstetrics and Gynecology, State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Ministry of Education, Key Laboratory of Assisted Reproduction (Peking University), Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- National Clinical Key Specialty Construction Program, Beijing, China
| |
Collapse
|
12
|
Chen X, Cheng G, Zhu L, Liu T, Yang X, Liu R, Ou Z, Zhang S, Tan W, Lin D, Wu C. Alarmin S100A8 imparts chemoresistance of esophageal cancer by reprogramming cancer-associated fibroblasts. Cell Rep Med 2024; 5:101576. [PMID: 38776909 PMCID: PMC11228400 DOI: 10.1016/j.xcrm.2024.101576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/08/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Chemotherapy remains the first-line treatment for advanced esophageal cancer. However, durable benefits are achieved by only a limited subset of individuals due to the elusive chemoresistance. Here, we utilize patient-derived xenografts (PDXs) from esophageal squamous-cell carcinoma to investigate chemoresistance mechanisms in preclinical settings. We observe that activated cancer-associated fibroblasts (CAFs) are enriched in the tumor microenvironment of PDXs resistant to chemotherapy. Mechanistically, we reveal that cancer-cell-derived S100A8 triggers the intracellular RhoA-ROCK-MLC2-MRTF-A pathway by binding to the CD147 receptor of CAFs, inducing CAF polarization and leading to chemoresistance. Therapeutically, we demonstrate that blocking the S100A8-CD147 pathway can improve chemotherapy efficiency. Prognostically, we found the S100A8 levels in peripheral blood can serve as an indicator of chemotherapy responsiveness. Collectively, our study offers a comprehensive understanding of the molecular mechanisms underlying chemoresistance in esophageal cancer and highlights the potential value of S100A8 in the clinical management of esophageal cancer.
Collapse
Affiliation(s)
- Xinjie Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Guoyu Cheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Liang Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Xinyu Yang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Rucheng Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Zhengjie Ou
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Wen Tan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing 100006, China.
| |
Collapse
|
13
|
Rezaei M, Mehta JL, Zadeh GM, Khedri A, Rezaei HB. Myosin light chain phosphatase is a downstream target of Rho-kinase in endothelin-1-induced transactivation of the TGF-β receptor. Cell Biochem Biophys 2024; 82:1109-1120. [PMID: 38834831 DOI: 10.1007/s12013-024-01262-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Rho-kinase (ROCK) regulates actomyosin contraction, coronary vasospasm, and cytoskeleton dynamics. ROCK and of NADPH oxidase (NOX) play an essential role in cardiovascular disease and proteoglycan synthesis, which promotes atherosclerosis by trapping low density lipoprotein. ROCK is activated by endothelin-1 (ET1) and transactivates the transforming growth factor beta receptor (TGFβR1), intensifying Smad signaling and proteoglycan production. This study aimed to identify the role of myosin light chain phosphatase (MLCP) as a downstream target of ROCK in TβR1 transactivation. METHODS Vascular smooth muscle cells were treated with ET1 and inhibitors of ROCK and MLCP were added. The phosphorylation levels of Smad2C, myosin light chain (MLC), and MLCP were monitored by western blot, and the mRNA expression of chondroitin 4-O-sulfotransferase 1 (C4ST1) was assessed by quantitative real-time PCR. RESULTS We examined ROCK's role in ET1-induced TGFβR1 activation. ROCK phosphorylated MLCP at the MYPT1 T853 residue, blocked by the ROCK inhibitor Y27632. ROCK also increased MLC phosphorylation and actomyosin contraction in response to ET1, enhanced by the phosphatase inhibitor Calyculin A. Calyculin A also increased C4ST1 expression, GAG-chain synthesizing enzymes. CONCLUSIONS This work suggests that ROCK is involved in ET1-mediated TβR1 activation through increased MLCP phosphorylation, which leads to Smad2C phosphorylation and stimulates C4ST1 expression.
Collapse
Affiliation(s)
- Maryam Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Jawahar Lal Mehta
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Ghorban Mohammad Zadeh
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Azam Khedri
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
14
|
Copeland I, Wonkam-Tingang E, Gupta-Malhotra M, Hashmi SS, Han Y, Jajoo A, Hall NJ, Hernandez PP, Lie N, Liu D, Xu J, Rosenfeld J, Haldipur A, Desire Z, Coban-Akdemir ZH, Scott DA, Li Q, Chao HT, Zaske AM, Lupski JR, Milewicz DM, Shete S, Posey JE, Hanchard NA. Exome sequencing implicates ancestry-related Mendelian variation at SYNE1 in childhood-onset essential hypertension. JCI Insight 2024; 9:e172152. [PMID: 38716726 PMCID: PMC11141928 DOI: 10.1172/jci.insight.172152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 03/19/2024] [Indexed: 05/12/2024] Open
Abstract
Childhood-onset essential hypertension (COEH) is an uncommon form of hypertension that manifests in childhood or adolescence and, in the United States, disproportionately affects children of African ancestry. The etiology of COEH is unknown, but its childhood onset, low prevalence, high heritability, and skewed ancestral demography suggest the potential to identify rare genetic variation segregating in a Mendelian manner among affected individuals and thereby implicate genes important to disease pathogenesis. However, no COEH genes have been reported to date. Here, we identify recessive segregation of rare and putatively damaging missense variation in the spectrin domain of spectrin repeat containing nuclear envelope protein 1 (SYNE1), a cardiovascular candidate gene, in 3 of 16 families with early-onset COEH without an antecedent family history. By leveraging exome sequence data from an additional 48 COEH families, 1,700 in-house trios, and publicly available data sets, we demonstrate that compound heterozygous SYNE1 variation in these COEH individuals occurred more often than expected by chance and that this class of biallelic rare variation was significantly enriched among individuals of African genetic ancestry. Using in vitro shRNA knockdown of SYNE1, we show that reduced SYNE1 expression resulted in a substantial decrease in the elasticity of smooth muscle vascular cells that could be rescued by pharmacological inhibition of the downstream RhoA/Rho-associated protein kinase pathway. These results provide insights into the molecular genetics and underlying pathophysiology of COEH and suggest a role for precision therapeutics in the future.
Collapse
Affiliation(s)
- Ian Copeland
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Edmond Wonkam-Tingang
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
| | | | - S. Shahrukh Hashmi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yixing Han
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
| | - Aarti Jajoo
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
| | - Nancy J. Hall
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- US Department of Agriculture Agricultural Research Service Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Paula P. Hernandez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- US Department of Agriculture Agricultural Research Service Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Natasha Lie
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
- US Department of Agriculture Agricultural Research Service Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Dan Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Jun Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Jill Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Baylor Genetics, Houston, Texas, USA
| | - Aparna Haldipur
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
| | - Zelene Desire
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
| | - Zeynep H. Coban-Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
- Department of Molecular Physiology and Biophysics
| | - Qing Li
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
| | - Hsiao-Tuan Chao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics; and
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital and Baylor College of Medicine, Houston, Texas, USA
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, Texas, USA
| | - Ana M. Zaske
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - James R. Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Dianna M. Milewicz
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sanjay Shete
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer E. Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, Texas, USA
| | - Neil A. Hanchard
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Childhood Complex Disease Genomics Section, National Human Genome Research Institute, NIH, Bethesda, USA
| |
Collapse
|
15
|
Dai Y, Zhang M, Liu X, Sun T, Qi W, Ding W, Chen Z, Zhang P, Liu R, Chen H, Chen S, Wang Y, Yue Y, Song N, Wang W, Jia H, Ma Z, Li C, Chen Q, Li B. Salmonella manipulates macrophage migration via SteC-mediated myosin light chain activation to penetrate the gut-vascular barrier. EMBO J 2024; 43:1499-1518. [PMID: 38528181 PMCID: PMC11021425 DOI: 10.1038/s44318-024-00076-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
The intestinal pathogen Salmonella enterica rapidly enters the bloodstream after the invasion of intestinal epithelial cells, but how Salmonella breaks through the gut-vascular barrier is largely unknown. Here, we report that Salmonella enters the bloodstream through intestinal CX3CR1+ macrophages during early infection. Mechanistically, Salmonella induces the migration/invasion properties of macrophages in a manner dependent on host cell actin and on the pathogen effector SteC. SteC recruits host myosin light chain protein Myl12a and phosphorylates its Ser19 and Thr20 residues. Myl12a phosphorylation results in actin rearrangement, and enhanced migration and invasion of macrophages. SteC is able to utilize a wide range of NTPs other than ATP to phosphorylate Myl12a. We further solved the crystal structure of SteC, which suggests an atypical dimerization-mediated catalytic mechanism. Finally, in vivo data show that SteC-mediated cytoskeleton manipulation is crucial for Salmonella breaching the gut vascular barrier and spreading to target organs.
Collapse
Affiliation(s)
- Yuanji Dai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Min Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoyu Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ting Sun
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wenqi Qi
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Ding
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhe Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Ping Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ruirui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Huimin Chen
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Siyan Chen
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuzhen Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yingying Yue
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Nannan Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haihong Jia
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhongrui Ma
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Cuiling Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Qixin Chen
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
| | - Bingqing Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
- Key Lab for Biotech-Drugs of National Health Commission, Jinan, 250117, China.
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, China.
| |
Collapse
|
16
|
Quintanilla MA, Patel H, Wu H, Sochacki KA, Chandrasekar S, Akamatsu M, Rotty JD, Korobova F, Bear JE, Taraska JW, Oakes PW, Beach JR. Local monomer levels and established filaments potentiate non-muscle myosin 2 assembly. J Cell Biol 2024; 223:e202305023. [PMID: 38353656 PMCID: PMC10866686 DOI: 10.1083/jcb.202305023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/02/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
The ability to dynamically assemble contractile networks is required throughout cell physiology, yet direct biophysical mechanisms regulating non-muscle myosin 2 filament assembly in living cells are lacking. Here, we use a suite of dynamic, quantitative imaging approaches to identify deterministic factors that drive myosin filament appearance and amplification. We find that actin dynamics regulate myosin assembly, but that the static actin architecture plays a less clear role. Instead, remodeling of actin networks modulates the local myosin monomer levels and facilitates assembly through myosin:myosin-driven interactions. Using optogenetically controlled myosin, we demonstrate that locally concentrating myosin is sufficient to both form filaments and jump-start filament amplification and partitioning. By counting myosin monomers within filaments, we demonstrate a myosin-facilitated assembly process that establishes filament stacks prior to partitioning into clusters that feed higher-order networks. Together, these findings establish the biophysical mechanisms regulating the assembly of non-muscle contractile structures that are ubiquitous throughout cell biology.
Collapse
Affiliation(s)
- Melissa A. Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Hiral Patel
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Huini Wu
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Kem A. Sochacki
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shreya Chandrasekar
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Matthew Akamatsu
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Jeremy D. Rotty
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Farida Korobova
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - James E. Bear
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Justin W. Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Patrick W. Oakes
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Jordan R. Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
17
|
Iyer KS, Maruri DP, Schmidtke DW, Petroll WM, Varner VD. Treatment with both TGF-β1 and PDGF-BB disrupts the stiffness-dependent myofibroblast differentiation of corneal keratocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582803. [PMID: 38496568 PMCID: PMC10942298 DOI: 10.1101/2024.02.29.582803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
During corneal wound healing, stromal keratocytes transform into a repair phenotype that is driven by the release of cytokines, like transforming growth factor-beta 1 (TGF-β1) and platelet-derived growth factor-BB (PDGF-BB). Previous work has shown that TGF-β1 promotes the myofibroblast differentiation of corneal keratocytes in a manner that depends on PDGF signaling. In addition, changes in mechanical properties are known to regulate the TGF-β1-mediated differentiation of cultured keratocytes. While PDGF signaling acts synergistically with TGF-β1 during myofibroblast differentiation, how treatment with multiple growth factors affects stiffness-dependent differences in keratocyte behavior is unknown. Here, we treated primary corneal keratocytes with PDGF-BB and TGF-β1 and cultured them on polyacrylamide (PA) substrata of different stiffnesses. In the presence of TGF-β1 alone, the cells underwent stiffness-dependent myofibroblast differentiation. On stiff substrata, the cells developed robust stress fibers, exhibited high levels of ⍺-SMA staining, formed large focal adhesions (FAs), and exerted elevated contractile forces, whereas cells in a compliant microenvironment showed low levels of ⍺-SMA immunofluorescence, formed smaller focal adhesions, and exerted decreased contractile forces. When the cultured keratocytes were treated simultaneously with PDGF-BB however, increased levels of ⍺-SMA staining and stress fiber formation were observed on compliant substrata, even though the cells did not exhibit elevated contractility or focal adhesion size. Pharmacological inhibition of PDGF signaling disrupted the myofibroblast differentiation of cells cultured on substrata of all stiffnesses. These results indicate that treatment with PDGF-BB can decouple molecular markers of myofibroblast differentiation from the elevated contractile phenotype otherwise associated with these cells, suggesting that crosstalk in the mechanotransductive signaling pathways downstream of TGF-β1 and PDGF-BB can regulate the stiffness-dependent differentiation of cultured keratocytes. Statement of Significance In vitro experiments have shown that changes in ECM stiffness can regulate the differentiation of myofibroblasts. Typically, these assays involve the use of individual growth factors, but it is unclear how stiffness-dependent differences in cell behavior are affected by multiple cytokines. Here, we used primary corneal keratocytes to show that treatment with both TGF-β1 and PDGF-BB disrupts the dependency of myofibroblast differentiation on substratum stiffness. In the presence of both growth factors, keratocytes on soft substrates exhibited elevated ⍺-SMA immunofluorescence without a corresponding increase in contractility or focal adhesion formation. This result suggests that molecular markers of myofibroblast differentiation can be dissociated from the elevated contractile behavior associated with the myofibroblast phenotype, suggesting potential crosstalk in mechanotransductive signaling pathways downstream of TGF-β1 and PDGF-BB.
Collapse
|
18
|
Nelson ZM, Leonard GD, Fehl C. Tools for investigating O-GlcNAc in signaling and other fundamental biological pathways. J Biol Chem 2024; 300:105615. [PMID: 38159850 PMCID: PMC10831167 DOI: 10.1016/j.jbc.2023.105615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
Cells continuously fine-tune signaling pathway proteins to match nutrient and stress levels in their local environment by modifying intracellular proteins with O-linked N-acetylglucosamine (O-GlcNAc) sugars, an essential process for cell survival and growth. The small size of these monosaccharide modifications poses a challenge for functional determination, but the chemistry and biology communities have together created a collection of precision tools to study these dynamic sugars. This review presents the major themes by which O-GlcNAc influences signaling pathway proteins, including G-protein coupled receptors, growth factor signaling, mitogen-activated protein kinase (MAPK) pathways, lipid sensing, and cytokine signaling pathways. Along the way, we describe in detail key chemical biology tools that have been developed and applied to determine specific O-GlcNAc roles in these pathways. These tools include metabolic labeling, O-GlcNAc-enhancing RNA aptamers, fluorescent biosensors, proximity labeling tools, nanobody targeting tools, O-GlcNAc cycling inhibitors, light-activated systems, chemoenzymatic labeling, and nutrient reporter assays. An emergent feature of this signaling pathway meta-analysis is the intricate interplay between O-GlcNAc modifications across different signaling systems, underscoring the importance of O-GlcNAc in regulating cellular processes. We highlight the significance of O-GlcNAc in signaling and the role of chemical and biochemical tools in unraveling distinct glycobiological regulatory mechanisms. Collectively, our field has determined effective strategies to probe O-GlcNAc roles in biology. At the same time, this survey of what we do not yet know presents a clear roadmap for the field to use these powerful chemical tools to explore cross-pathway O-GlcNAc interactions in signaling and other major biological pathways.
Collapse
Affiliation(s)
- Zachary M Nelson
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Garry D Leonard
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Charlie Fehl
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA.
| |
Collapse
|
19
|
Futterknecht S, Chatzimichail E, Gugleta K, Panos GD, Gatzioufas Z. The Role of Rho Kinase Inhibitors in Corneal Diseases. Drug Des Devel Ther 2024; 18:97-108. [PMID: 38264539 PMCID: PMC10804875 DOI: 10.2147/dddt.s435522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024] Open
Abstract
The cornea, as the outermost layer of the eye, plays a crucial role in vision by focusing light onto the retina. Various diseases and injuries can compromise its clarity, leading to impaired vision. This review aims to provide a thorough overview of the pharmacological properties, therapeutic potential and associated risks of Rho-associated protein kinase (ROCK) inhibitors in the management of corneal diseases. The article focuses on four key ROCK inhibitors: Y-27632, fasudil, ripasudil, and netarsudil, providing a comparative examination. Studies supporting the use of ROCK inhibitors highlight their efficacy across diverse corneal conditions. In Fuchs' endothelial corneal dystrophy, studies on the application of Y-27632, ripasudil, and netarsudil demonstrated noteworthy enhancements in corneal clarity, endothelial cell density, and visual acuity. In pseudophakic bullous keratopathy, the injection of Y-27632 together with cultured corneal endothelial cells into the anterior chamber lead to enhanced corneal endothelial cell density and improved visual acuity. Animal models simulating chemical injury to the cornea showed a reduction of neovascularization and epithelial defects after application of fasudil and in a case of iridocorneal endothelial syndrome netarsudil improved corneal edema. Addressing safety considerations, netarsudil and ripasudil, both clinically approved, exhibit adverse events such as conjunctival hyperemia, conjunctival hemorrhage, cornea verticillata, conjunctivitis, and blepharitis. Monitoring patients during treatment becomes crucial to balancing the potential therapeutic benefits with these associated risks. In conclusion, ROCK inhibitors, particularly netarsudil and ripasudil, offer promise in managing corneal diseases. The comparative analysis of their pharmacological properties and studies supporting their efficacy underscore their potential therapeutic significance. However, ongoing research is paramount to comprehensively understand their safety profiles and long-term outcomes in diverse corneal conditions, guiding their optimal application in clinical practice.
Collapse
Affiliation(s)
- Stefan Futterknecht
- Department of Ophthalmology, University Hospital of Basel, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | | | - Konstantin Gugleta
- Department of Ophthalmology, University Hospital of Basel, Basel, Switzerland
- Department of Ophthalmology, School of Medicine, University of Basel, Basel, Switzerland
| | - Georgios D Panos
- Department of Ophthalmology, Queen’s Medical Centre, Nottingham University Hospitals, Nottingham, UK
- Division of Ophthalmology and Visual Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Zisis Gatzioufas
- Department of Ophthalmology, University Hospital of Basel, Basel, Switzerland
- Department of Ophthalmology, School of Medicine, University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
Gidaro A, Delitala AP, Manetti R, Caccia S, Soloski MJ, Lambertenghi Deliliers G, Castro D, Donadoni M, Bartoli A, Sanna G, Bergamaschini L, Castelli R. Platelet Microvesicles, Inflammation, and Coagulation Markers: A Pilot Study. Hematol Rep 2023; 15:684-695. [PMID: 38132277 PMCID: PMC10742513 DOI: 10.3390/hematolrep15040069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/31/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Platelet "Microvesicles" (MVs) are studied for their role in blood coagulation and inflammation. The study aimed to establish if MVs are related to age, plasma levels of inflammation, coagulation, and fibrinolysis markers in healthy individuals. METHODS We prospectively enrolled volunteers aged over 18 years. MVs, plasma levels of C-reactive protein (CRP), Interleukin 6 (IL-6), Interleukin 10 (IL-10), Interleukin 17 (IL-17), and transforming growth factor β (TGF-β), fibrinogen, plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor (VWF), homocysteine, factor VII (FVII), thrombin activatable fibrinolysis inhibitor (TAFI), and Protein S were tested. RESULTS A total of 246 individuals (median age 65 years ("IQR"54-72)) were evaluated. Both univariate analysis and logistic regression models showed that MVs positively correlate with age, CRP, IL-6, IL-10, IL-17, TGF-β, fibrinogen, PAI-1, VWF, FVII, and homocysteine, while inversely correlating with TAFI and Protein S. The ROC curve analysis performed to identify a cut off for MV values (700 kMP) showed a good accuracy with over-range cytokines fibrinolysis factor and coagulation markers. CONCLUSIONS To the best of our knowledge, this study is the first to correlate MVs with an entire panel of cardiovascular risk factors in healthy individuals. A future possible role of MVs in screening exams is suggested.
Collapse
Affiliation(s)
- Antonio Gidaro
- Department of Biomedical and Clinical Sciences Luigi Sacco, Luigi Sacco Hospital, University of Milan, Via G.B. Grassi N° 74, 20157 Milan, Italy; (S.C.); (M.D.); (A.B.); (L.B.)
| | - Alessandro Palmerio Delitala
- Department of Medicine, Surgery and Pharmacy University of Sassari, Via San Pietro 43, 07100 Sassari, Italy; (A.P.D.); (R.M.); (D.C.); (G.S.)
| | - Roberto Manetti
- Department of Medicine, Surgery and Pharmacy University of Sassari, Via San Pietro 43, 07100 Sassari, Italy; (A.P.D.); (R.M.); (D.C.); (G.S.)
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences Luigi Sacco, Luigi Sacco Hospital, University of Milan, Via G.B. Grassi N° 74, 20157 Milan, Italy; (S.C.); (M.D.); (A.B.); (L.B.)
| | - Mark J. Soloski
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA;
| | | | - Dante Castro
- Department of Medicine, Surgery and Pharmacy University of Sassari, Via San Pietro 43, 07100 Sassari, Italy; (A.P.D.); (R.M.); (D.C.); (G.S.)
| | - Mattia Donadoni
- Department of Biomedical and Clinical Sciences Luigi Sacco, Luigi Sacco Hospital, University of Milan, Via G.B. Grassi N° 74, 20157 Milan, Italy; (S.C.); (M.D.); (A.B.); (L.B.)
| | - Arianna Bartoli
- Department of Biomedical and Clinical Sciences Luigi Sacco, Luigi Sacco Hospital, University of Milan, Via G.B. Grassi N° 74, 20157 Milan, Italy; (S.C.); (M.D.); (A.B.); (L.B.)
| | - Giuseppe Sanna
- Department of Medicine, Surgery and Pharmacy University of Sassari, Via San Pietro 43, 07100 Sassari, Italy; (A.P.D.); (R.M.); (D.C.); (G.S.)
| | - Luigi Bergamaschini
- Department of Biomedical and Clinical Sciences Luigi Sacco, Luigi Sacco Hospital, University of Milan, Via G.B. Grassi N° 74, 20157 Milan, Italy; (S.C.); (M.D.); (A.B.); (L.B.)
| | - Roberto Castelli
- Department of Medicine, Surgery and Pharmacy University of Sassari, Via San Pietro 43, 07100 Sassari, Italy; (A.P.D.); (R.M.); (D.C.); (G.S.)
| |
Collapse
|
21
|
Li R, Feng D, Han S, Zhai X, Yu X, Fu Y, Jin F. Macrophages and fibroblasts in foreign body reactions: How mechanical cues drive cell functions? Mater Today Bio 2023; 22:100783. [PMID: 37701130 PMCID: PMC10494263 DOI: 10.1016/j.mtbio.2023.100783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Biomaterials, when implanted in the human body, can induce a series of cell- and cytokine-related reactions termed foreign body reactions (FBRs). In the progression of FBRs, macrophages regulate inflammation and healing by polarizing to either a pro-inflammatory or pro-healing phenotype and recruit fibroblasts by secreting cytokines. Stimulated by the biomaterials, fibrotic capsule is formed eventually. The implant, along with its newly formed capsule, introduces various mechanical cues that influence cellular functions. Mechanosensing proteins, such as integrins or ion channels, transduce extracellular mechanical signals into cytoplasm biochemical signals in response to mechanical stimuli. Consequently, the morphology, migration mode, function, and polarization state of the cells are affected. Modulated by different intracellular signaling pathways and their crosstalk, the expression of fibrotic genes increases with fibroblast activation and fibroblast to myofibroblast transition under stiff or force stimuli. However, summarized in most current studies, the outcomes of macrophage polarization in the effect of different mechanical cues are inconsistent. The underlying mechanisms should be investigated with more advanced technology and considering more interfering aspects. Further research is needed to determine how to modulate the progression of fibrotic capsule formation in FBR artificially.
Collapse
Affiliation(s)
- Rihan Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Dongdong Feng
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Siyuan Han
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Xiaoyue Zhai
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110000, China
| | - Xinmiao Yu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
- Department of Breast and Reconstructive Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Yuanyuan Fu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110000, China
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| |
Collapse
|
22
|
Wang Y, Troughton LD, Xu F, Chatterjee A, Ding C, Zhao H, Cifuentes LP, Wagner RB, Wang T, Tan S, Chen J, Li L, Umulis D, Kuang S, Suter DM, Yuan C, Chan D, Huang F, Oakes PW, Deng Q. Atypical peripheral actin band formation via overactivation of RhoA and nonmuscle myosin II in mitofusin 2-deficient cells. eLife 2023; 12:e88828. [PMID: 37724949 PMCID: PMC10550287 DOI: 10.7554/elife.88828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/19/2023] [Indexed: 09/21/2023] Open
Abstract
Cell spreading and migration play central roles in many physiological and pathophysiological processes. We have previously shown that MFN2 regulates the migration of human neutrophil-like cells via suppressing Rac activation. Here, we show that in mouse embryonic fibroblasts, MFN2 suppresses RhoA activation and supports cell polarization. After initial spreading, the wild-type cells polarize and migrate, whereas the Mfn2-/- cells maintain a circular shape. Increased cytosolic Ca2+ resulting from the loss of Mfn2 is directly responsible for this phenotype, which can be rescued by expressing an artificial tether to bring mitochondria and endoplasmic reticulum to close vicinity. Elevated cytosolic Ca2+ activates Ca2+/calmodulin-dependent protein kinase II, RhoA, and myosin light-chain kinase, causing an overactivation of nonmuscle myosin II, leading to a formation of a prominent F-actin ring at the cell periphery and increased cell contractility. The peripheral actin band alters cell physics and is dependent on substrate rigidity. Our results provide a novel molecular basis to understand how MFN2 regulates distinct signaling pathways in different cells and tissue environments, which is instrumental in understanding and treating MFN2-related diseases.
Collapse
Affiliation(s)
- Yueyang Wang
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Lee D Troughton
- Cell and Molecular Physiology, Loyola University ChicagoChicagoUnited States
| | - Fan Xu
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of TechnologyBeijingChina
| | - Aritra Chatterjee
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Chang Ding
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Han Zhao
- Davidson School of Chemical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Laura P Cifuentes
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Ryan B Wagner
- School of Mechanical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Tianqi Wang
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Shelly Tan
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Jingjuan Chen
- Department of Animal Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Linlin Li
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - David Umulis
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
- Department of Agricultural and Biological Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
- Purdue Institute for Integrative Neuroscience, Purdue University West LafayetteWest LafayetteUnited States
- Purdue Institute for Inflammation, Immunology & Infectious Disease, Purdue University West LafayetteWest LafayetteUnited States
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Deva Chan
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Fang Huang
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Patrick W Oakes
- Cell and Molecular Physiology, Loyola University ChicagoChicagoUnited States
| | - Qing Deng
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
- Purdue Institute for Inflammation, Immunology & Infectious Disease, Purdue University West LafayetteWest LafayetteUnited States
- Purdue University Center for Cancer Research, Purdue University West LafayetteWest LafayetteUnited States
| |
Collapse
|
23
|
Wang C, Du M, Jiang Z, Cong R, Wang W, Zhang G, Li L. Comparative proteomic and phosphoproteomic analysis reveals differential heat response mechanism in two congeneric oyster species. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115197. [PMID: 37451098 DOI: 10.1016/j.ecoenv.2023.115197] [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: 04/29/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023]
Abstract
High-temperature stress caused by global climate change poses a significant threat to marine ectotherms. This study investigated the role of protein phosphorylation modifications in the molecular regulation network under heat stress in oysters, which are representative intertidal organisms that experience considerable temperature changes. Firstly, the study compared the extent of thermal damage between two congeneric oyster species, the relative heat-tolerant Crassostrea angulata (C. angulata) and heat-sensitive Crassostrea gigas (C. gigas), under sublethal temperature (37 °C) for 12 h, using various physiological and biochemical methods. Subsequently, the comparative proteomic and phosphoproteomic analyses revealed that high-temperature considerably regulated signal transduction, energy metabolism, protein synthesis, cell survival and apoptosis, and cytoskeleton remodeling through phosphorylation modifications of related receptors and kinases. Furthermore, the protein kinase A, mitogen-activated protein kinase 1, tyrosine-protein kinase Src, and serine/threonine kinase AKT, exhibiting differential phosphorylation modification patterns, were identified as hub regulators that may enhance glycolysis and TCA cycle to increase the energy supply, distribute protein synthesis, inhibit Caspase-dependent apoptosis activated by endogenous mitochondrial cytochrome release and maintain cytoskeletal stability, ultimately shaping the higher thermal resistance of C. angulata. This study represents the first investigation of protein phosphorylation dynamics in marine invertebrates under heat stress, reveals the molecular mechanisms underlying the differential thermal responses between two Crassostrea oysters at the phosphorylation level, and provides new insights into understanding phosphorylation-mediated molecular responses in marine organisms during environmental changes and predicting the adaptive potential in the context of global warming.
Collapse
Affiliation(s)
- Chaogang Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China
| | - Mingyang Du
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhuxiang Jiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China
| | - Rihao Cong
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Wei Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Guofan Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Li Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China.
| |
Collapse
|
24
|
Liu W, Lu JY, Wang YJ, Xu XX, Chen YC, Yu SX, Xiang XW, Chen XZ, Jiu Y, Gao H, Sheng M, Chen ZJ, Hu X, Li D, Maiuri P, Huang X, Ying T, Xu GL, Pang DW, Zhang ZL, Liu B, Liu YJ. Vaccinia virus induces EMT-like transformation and RhoA-mediated mesenchymal migration. J Med Virol 2023; 95:e29041. [PMID: 37621182 DOI: 10.1002/jmv.29041] [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: 05/24/2023] [Revised: 07/17/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
The emerging outbreak of monkeypox is closely associated with the viral infection and spreading, threatening global public health. Virus-induced cell migration facilitates viral transmission. However, the mechanism underlying this type of cell migration remains unclear. Here we investigate the motility of cells infected by vaccinia virus (VACV), a close relative of monkeypox, through combining multi-omics analyses and high-resolution live-cell imaging. We find that, upon VACV infection, the epithelial cells undergo epithelial-mesenchymal transition-like transformation, during which they lose intercellular junctions and acquire the migratory capacity to promote viral spreading. After transformation, VACV-hijacked RhoA signaling significantly alters cellular morphology and rearranges the actin cytoskeleton involving the depolymerization of robust actin stress fibers, leading-edge protrusion formation, and the rear-edge recontraction, which coordinates VACV-induced cell migration. Our study reveals how poxviruses alter the epithelial phenotype and regulate RhoA signaling to induce fast migration, providing a unique perspective to understand the pathogenesis of poxviruses.
Collapse
Affiliation(s)
- Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Jia-Yin Lu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Xin-Xin Xu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Yu-Chen Chen
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Xiao-Wei Xiang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Xue-Zhu Chen
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Yaming Jiu
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Hai Gao
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Mengyao Sheng
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Zheng-Jun Chen
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xinyao Hu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, College of Life Sciences, Institute of Biophysics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Dong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, College of Life Sciences, Institute of Biophysics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Paolo Maiuri
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Xinxin Huang
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Guo-Liang Xu
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, Frontiers Science Center for Cell Responses, College of Chemistry, Nankai University, Tianjin, China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Baohong Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Woodbury SM, Swanson WB, Mishina Y. Mechanobiology-informed biomaterial and tissue engineering strategies for influencing skeletal stem and progenitor cell fate. Front Physiol 2023; 14:1220555. [PMID: 37520820 PMCID: PMC10373313 DOI: 10.3389/fphys.2023.1220555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023] Open
Abstract
Skeletal stem and progenitor cells (SSPCs) are the multi-potent, self-renewing cell lineages that form the hematopoietic environment and adventitial structures of the skeletal tissues. Skeletal tissues are responsible for a diverse range of physiological functions because of the extensive differentiation potential of SSPCs. The differentiation fates of SSPCs are shaped by the physical properties of their surrounding microenvironment and the mechanical loading forces exerted on them within the skeletal system. In this context, the present review first highlights important biomolecules involved with the mechanobiology of how SSPCs sense and transduce these physical signals. The review then shifts focus towards how the static and dynamic physical properties of microenvironments direct the biological fates of SSPCs, specifically within biomaterial and tissue engineering systems. Biomaterial constructs possess designable, quantifiable physical properties that enable the growth of cells in controlled physical environments both in-vitro and in-vivo. The utilization of biomaterials in tissue engineering systems provides a valuable platform for controllably directing the fates of SSPCs with physical signals as a tool for mechanobiology investigations and as a template for guiding skeletal tissue regeneration. It is paramount to study this mechanobiology and account for these mechanics-mediated behaviors to develop next-generation tissue engineering therapies that synergistically combine physical and chemical signals to direct cell fate. Ultimately, taking advantage of the evolved mechanobiology of SSPCs with customizable biomaterial constructs presents a powerful method to predictably guide bone and skeletal organ regeneration.
Collapse
Affiliation(s)
- Seth M. Woodbury
- Yuji Mishina Laboratory, University of Michigan School of Dentistry, Department of Biologic and Materials Science & Prosthodontics, Ann Arbor, MI, United States
- University of Michigan College of Literature, Science, and Arts, Department of Chemistry, Ann Arbor, MI, United States
- University of Michigan College of Literature, Science, and Arts, Department of Physics, Ann Arbor, MI, United States
| | - W. Benton Swanson
- Yuji Mishina Laboratory, University of Michigan School of Dentistry, Department of Biologic and Materials Science & Prosthodontics, Ann Arbor, MI, United States
| | - Yuji Mishina
- Yuji Mishina Laboratory, University of Michigan School of Dentistry, Department of Biologic and Materials Science & Prosthodontics, Ann Arbor, MI, United States
| |
Collapse
|
26
|
Liu J, Guo Y, Zhang R, Xu Y, Luo C, Wang R, Xu S, Wei L. Inhibition of TRPV4 remodels single cell polarity and suppresses the metastasis of hepatocellular carcinoma. Cell Death Dis 2023; 14:379. [PMID: 37369706 DOI: 10.1038/s41419-023-05903-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 05/31/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor, frequently causing both intrahepatic and extrahepatic metastases. The overall prognosis of patients with metastatic HCC is poor. Recently, single-cell (sc) polarity is proved to be an innate feature of some tumor cells in liquid phase, and directly involved in the cell adhesion to blood vessel and tumor metastasis. Here, we characterize the maintained sc polarity of HCC cells in a suspension culture, and investigate its roles and regulatory mechanisms during metastasis. We demonstrate that transient receptor potential vanilloid 4 (TRPV4) is a promoting regulator of sc polarity via activating Ca2+-dependent AMPK/MLC/ERM pathway. This attenuates the adhesion of metastatic HCC cells to vascular endothelial cells. The reduction of cancer metastases can result from TRPV4 inhibition, which not only impacts the migration and invasion of tumor cells, but also prevents the adhesion to vascular endothelial cells. Additionally, we discover a brand-new TRPV4 inhibitor called GL-V9 that modifies the degree of sc polarization and significantly decreases the metastatic capacity of HCC cells. Taken together, our data shows that TRPV4 and calcium signal are significant sc polarity regulators in metastatic HCC, and that the pharmacological intervention that results in HCC cells becoming depolarized suggests a promising treatment for cancer metastasis.
Collapse
Affiliation(s)
- Jian Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, #24 Tongjiaxiang, Nanjing, The People's Republic of China
| | - Yongjian Guo
- School of Biopharmacy, China Pharmaceutical University, #639 Longmian Dadao, Nanjing, The People's Republic of China
| | - Ruitian Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, #24 Tongjiaxiang, Nanjing, The People's Republic of China
| | - Ye Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, #24 Tongjiaxiang, Nanjing, The People's Republic of China
| | - Chengju Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, #24 Tongjiaxiang, Nanjing, The People's Republic of China
| | - Rui Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, #24 Tongjiaxiang, Nanjing, The People's Republic of China
| | - Shu Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, #24 Tongjiaxiang, Nanjing, The People's Republic of China.
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, #24 Tongjiaxiang, Nanjing, The People's Republic of China.
| |
Collapse
|
27
|
Yan R, Liu D, Wang J, Liu M, Guo H, Bai J, Yang S, Chang J, Yao Z, Yang Z, Blom T, Zhou K. miR-137-LAPTM4B regulates cytoskeleton organization and cancer metastasis via the RhoA-LIMK-Cofilin pathway in osteosarcoma. Oncogenesis 2023; 12:25. [PMID: 37147294 PMCID: PMC10163001 DOI: 10.1038/s41389-023-00471-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/07/2023] Open
Abstract
Osteosarcoma (OS) is a rare malignant bone tumor but is one leading cause of cancer mortality in childhood and adolescence. Cancer metastasis accounts for the primary reason for treatment failure in OS patients. The dynamic organization of the cytoskeleton is fundamental for cell motility, migration, and cancer metastasis. Lysosome Associated Protein Transmembrane 4B (LAPTM4B) is an oncogene participating in various biological progress central to cancer biogenesis. However, the potential roles of LAPTM4B in OS and the related mechanisms remain unknown. Here, we established the elevated LAPTM4B expression in OS, and it is essential in regulating stress fiber organization through RhoA-LIMK-cofilin signaling pathway. In terms of mechanism, our data revealed that LAPTM4B promotes RhoA protein stability by suppressing the ubiquitin-mediated proteasome degradation pathway. Moreover, our data show that miR-137, rather than gene copy number and methylation status, contributes to the upregulation of LAPTM4B in OS. We report that miR-137 is capable of regulating stress fiber arrangement, OS cell migration, and metastasis via targeting LAPTM4B. Combining results from cells, patients' tissue samples, the animal model, and cancer databases, this study further suggests that the miR-137-LAPTM4B axis represents a clinically relevant pathway in OS progression and a viable target for novel therapeutics.
Collapse
Affiliation(s)
- Ruyu Yan
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Dan Liu
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Junjie Wang
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Minxia Liu
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, 00290, Finland
| | - Hongjuan Guo
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jing Bai
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shuo Yang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Jun Chang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhihong Yao
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Tomas Blom
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, 00014, Finland.
| | - Kecheng Zhou
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, 00014, Finland.
| |
Collapse
|
28
|
Quintanilla MA, Patel H, Wu H, Sochacki KA, Akamatsu M, Rotty JD, Korobova F, Bear JE, Taraska JW, Oakes PW, Beach JR. Local Monomer Levels and Established Filaments Potentiate Non-Muscle Myosin 2 Assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538303. [PMID: 37162845 PMCID: PMC10168331 DOI: 10.1101/2023.04.26.538303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The ability to dynamically assemble contractile networks is required throughout cell physiology, yet the biophysical mechanisms regulating non-muscle myosin 2 filament assembly in living cells are lacking. Here we use a suite of dynamic, quantitative imaging approaches to identify deterministic factors that drive myosin filament appearance and amplification. We find that actin dynamics regulate myosin assembly, but that the actin architecture plays a minimal direct role. Instead, remodeling of actin networks modulates the local myosin monomer levels and facilitates assembly through myosin:myosin driven interactions. Using optogenetically controlled myosin, we demonstrate that locally concentrating myosin is sufficient to both form filaments and jump-start filament amplification and partitioning. By counting myosin monomers within filaments, we demonstrate a myosin-facilitated assembly process that establishes sub-resolution filament stacks prior to partitioning into clusters that feed higher-order networks. Together these findings establish the biophysical mechanisms regulating the assembly of non-muscle contractile structures that are ubiquitous throughout cell biology.
Collapse
Affiliation(s)
- Melissa A Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Hiral Patel
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Huini Wu
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Kem A Sochacki
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | | | - Jeremy D Rotty
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Farida Korobova
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Patrick W Oakes
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Jordan R Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| |
Collapse
|
29
|
Lei Y, Fu X, Chen M, Yi Y, Mao P, Peng L, Qu Z. Dahuang—Taoren, a botanical drug combination, ameliorates adenomyosis via inhibiting Rho GTPases. Front Pharmacol 2023; 14:1089004. [PMID: 36969843 PMCID: PMC10035534 DOI: 10.3389/fphar.2023.1089004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Introduction: Dahuang-Taoren (DT) is a classic combination of botanical drugs applied to treat pain-related diseases in ancient China. Today, DT is frequently applied for dysmenorrhea of adenomyosis (AM) in the clinic. Growing evidence indicates Rho GTPases may play an essential role in AM progression. However, the potential mechanism of DT on Rho GTPases in AM remains unclear.Methods: The expressions of Rho GTPases in the patients with AM were evaluated. Further, pituitary transplantation-induced AM mice and the primary AM endometrial stromal cells (AMESCs) were subjected to DT intervention.Results: The results revealed that the expressions of Rho GTPases were significantly upregulated in both AM patients and AM mice. The DT could reduce pathological infiltration, relieve hyperalgesia, and alleviate cytoskeleton remodeling in AM mice. Besides, the migration and invasion of AMESCs were markedly inhibited after exposure to DT.Discussion: These effects may be linked to the decreased Rho GTPases expression. The results may offer a novel explanation of DT against AM.
Collapse
Affiliation(s)
- Ya Lei
- The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People’s Hospital, Yichang, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Xianyun Fu
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Minmin Chen
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Yongli Yi
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Ping Mao
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
| | - Li Peng
- The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People’s Hospital, Yichang, China
- *Correspondence: Li Peng, ; Zhao Qu,
| | - Zhao Qu
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, China
- *Correspondence: Li Peng, ; Zhao Qu,
| |
Collapse
|
30
|
Hou Y, Li Y, Li Y, Li D, Guo T, Deng X, Zhang H, Xie C, Lu X. Tuning Water-Resistant Networks in Mussel-Inspired Hydrogels for Robust Wet Tissue and Bioelectronic Adhesion. ACS NANO 2023; 17:2745-2760. [PMID: 36734875 DOI: 10.1021/acsnano.2c11053] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Hydrogels with robust wet adhesion are desirable for applications in aqueous environments. Wet adhesion arising from synergy between hydrophobic and catechol components in mussel foot proteins has been highlighted. However, optimizing hydrogels with multiple components is challenging because of their complex structure-property relationships. Herein, high-throughput screening of a series of hydrophobic alkyl monomers and adhesive catechol derivatives was used to systematically develop wet adhesive hydrogels. Short alkyl chains promote wet adhesion by repelling water at the adhesive interface, whereas long alkyl chains form strong hydrophobic interactions inside the hydrogel network that impede or dissipate energy for wet adhesion. The optimized wet adhesive hydrogel, containing short alkyl chain, was applied for rapid hemostasis and wound healing because of the synergistic effect of catechol and alkyl groups and its immunomodulation ability, which is revealed through a transcriptomic analysis. Conductive nanocomponents were incorporated into the optimized hydrogel to produce a wearable device, which was used for continuous monitoring human electrocardiogram (ECG) during swimming, and in situ epicardial ECG on a porcine living and beating heart. This study demonstrated an efficient and generalized molecular design strategy for multifunctional wet adhesive hydrogels.
Collapse
Affiliation(s)
- Yue Hou
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yazhen Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai 200125, China
| | - Yingqi Li
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Da Li
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tailin Guo
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Xu Deng
- Institute of Fundamental and Frontier Science, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hongping Zhang
- School of Mechanical Engineering, Institute for Advanced Study, Chengdu University, Chengdu, Sichuan 610041, China
| | - Chaoming Xie
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiong Lu
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
31
|
Guan G, Cannon RD, Coates DE, Mei L. Effect of the Rho-Kinase/ROCK Signaling Pathway on Cytoskeleton Components. Genes (Basel) 2023; 14:272. [PMID: 36833199 PMCID: PMC9957420 DOI: 10.3390/genes14020272] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The mechanical properties of cells are important in tissue homeostasis and enable cell growth, division, migration and the epithelial-mesenchymal transition. Mechanical properties are determined to a large extent by the cytoskeleton. The cytoskeleton is a complex and dynamic network composed of microfilaments, intermediate filaments and microtubules. These cellular structures confer both cell shape and mechanical properties. The architecture of the networks formed by the cytoskeleton is regulated by several pathways, a key one being the Rho-kinase/ROCK signaling pathway. This review describes the role of ROCK (Rho-associated coiled-coil forming kinase) and how it mediates effects on the key components of the cytoskeleton that are critical for cell behaviour.
Collapse
Affiliation(s)
- Guangzhao Guan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Diagnostic and Surgical Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Richard D. Cannon
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Dawn E. Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Li Mei
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| |
Collapse
|
32
|
Jayarajan V, Hall GT, Xenakis T, Bulstrode N, Moulding D, Castellano S, Di WL. Short-Term Treatment with Rho-Associated Kinase Inhibitor Preserves Keratinocyte Stem Cell Characteristics In Vitro. Cells 2023; 12:cells12030346. [PMID: 36766688 PMCID: PMC9913223 DOI: 10.3390/cells12030346] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Primary keratinocytes including keratinocyte stem cells (KSCs) can be cultured as epidermal sheets in vitro and are attractive for cell and gene therapies for genetic skin disorders. However, the initial slow growth of freshly isolated keratinocytes hinders clinical applications. Rho-associated kinase inhibitor (ROCKi) has been used to overcome this obstacle, but its influence on the characteristics of KSC and its safety for clinical application remains unknown. In this study, primary keratinocytes were treated with ROCKi Y-27632 for six days (short-term). Significant increases in colony formation and cell proliferation during the six-day ROCKi treatment were observed and confirmed by related protein markers and single-cell transcriptomic analysis. In addition, short-term ROCKi-treated cells maintained their differentiation ability as examined by 3D-organotypic culture. However, these changes could be reversed and became indistinguishable between treated and untreated cells once ROCKi treatment was withdrawn. Further, the short-term ROCKi treatment did not reduce the number of KSCs. In addition, AKT and ERK pathways were rapidly activated upon ROCKi treatment. In conclusion, short-term ROCKi treatment can transiently and reversibly accelerate initial primary keratinocyte expansion while preserving the holoclone-forming cell population (KSCs), providing a safe avenue for clinical applications.
Collapse
Affiliation(s)
- Vignesh Jayarajan
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - George T. Hall
- Genetics and Genomic Medicine Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, 20 Guilford Street, London WC1N 1DZ, UK
| | - Theodoros Xenakis
- Genetics and Genomic Medicine Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, 20 Guilford Street, London WC1N 1DZ, UK
| | - Neil Bulstrode
- Department of Plastic Surgery, Great Ormond Street Hospital for Children, Great Ormond Street, London WC1N 3JH, UK
| | - Dale Moulding
- Light Microscopy Core Facility, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Sergi Castellano
- Genetics and Genomic Medicine Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, 20 Guilford Street, London WC1N 1DZ, UK
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, 20 Guilford Street, London WC1N 1DZ, UK
| | - Wei-Li Di
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- Correspondence: ; Tel.: +44-(0)207905-2369; Fax: +44-(0)207905-2882
| |
Collapse
|
33
|
Aguilar VM, Paul A, Lazarko D, Levitan I. Paradigms of endothelial stiffening in cardiovascular disease and vascular aging. Front Physiol 2023; 13:1081119. [PMID: 36714307 PMCID: PMC9874005 DOI: 10.3389/fphys.2022.1081119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Endothelial cells, the inner lining of the blood vessels, are well-known to play a critical role in vascular function, while endothelial dysfunction due to different cardiovascular risk factors or accumulation of disruptive mechanisms that arise with aging lead to cardiovascular disease. In this review, we focus on endothelial stiffness, a fundamental biomechanical property that reflects cell resistance to deformation. In the first part of the review, we describe the mechanisms that determine endothelial stiffness, including RhoA-dependent contractile response, actin architecture and crosslinking, as well as the contributions of the intermediate filaments, vimentin and lamin. Then, we review the factors that induce endothelial stiffening, with the emphasis on mechanical signals, such as fluid shear stress, stretch and stiffness of the extracellular matrix, which are well-known to control endothelial biomechanics. We also describe in detail the contribution of lipid factors, particularly oxidized lipids, that were also shown to be crucial in regulation of endothelial stiffness. Furthermore, we discuss the relative contributions of these two mechanisms of endothelial stiffening in vasculature in cardiovascular disease and aging. Finally, we present the current state of knowledge about the role of endothelial stiffening in the disruption of endothelial cell-cell junctions that are responsible for the maintenance of the endothelial barrier.
Collapse
Affiliation(s)
- Victor M. Aguilar
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Amit Paul
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Dana Lazarko
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
34
|
Ojasalu K, Lieber S, Sokol AM, Nist A, Stiewe T, Bullwinkel I, Finkernagel F, Reinartz S, Müller-Brüsselbach S, Grosse R, Graumann J, Müller R. The lysophosphatidic acid-regulated signal transduction network in ovarian cancer cells and its role in actomyosin dynamics, cell migration and entosis. Theranostics 2023; 13:1921-1948. [PMID: 37064875 PMCID: PMC10091871 DOI: 10.7150/thno.81656] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/25/2023] [Indexed: 04/18/2023] Open
Abstract
Lysophosphatidic acid (LPA) species accumulate in the ascites of ovarian high-grade serous cancer (HGSC) and are associated with short relapse-free survival. LPA is known to support metastatic spread of cancer cells by activating a multitude of signaling pathways via G-protein-coupled receptors of the LPAR family. Systematic unbiased analyses of the LPA-regulated signal transduction network in ovarian cancer cells have, however, not been reported to date. Methods: LPA-induced signaling pathways were identified by phosphoproteomics of both patient-derived and OVCAR8 cells, RNA sequencing, measurements of intracellular Ca2+ and cAMP as well as cell imaging. The function of LPARs and downstream signaling components in migration and entosis were analyzed by selective pharmacological inhibitors and RNA interference. Results: Phosphoproteomic analyses identified > 1100 LPA-regulated sites in > 800 proteins and revealed interconnected LPAR1, ROCK/RAC, PKC/D and ERK pathways to play a prominent role within a comprehensive signaling network. These pathways regulate essential processes, including transcriptional responses, actomyosin dynamics, cell migration and entosis. A critical component of this signaling network is MYPT1, a stimulatory subunit of protein phosphatase 1 (PP1), which in turn is a negative regulator of myosin light chain 2 (MLC2). LPA induces phosphorylation of MYPT1 through ROCK (T853) and PKC/ERK (S507), which is majorly driven by LPAR1. Inhibition of MYPT1, PKC or ERK impedes both LPA-induced cell migration and entosis, while interference with ROCK activity and MLC2 phosphorylation selectively blocks entosis, suggesting that MYPT1 figures in both ROCK/MLC2-dependent and -independent pathways. We finally show a novel pathway governed by LPAR2 and the RAC-GEF DOCK7 to be indispensable for the induction of entosis. Conclusion: We have identified a comprehensive LPA-induced signal transduction network controlling LPA-triggered cytoskeletal changes, cell migration and entosis in HGSC cells. Due to its pivotal role in this network, MYPT1 may represent a promising target for interfering with specific functions of PP1 essential for HGSC progression.
Collapse
Affiliation(s)
- Kaire Ojasalu
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Sonja Lieber
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Anna M. Sokol
- Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Imke Bullwinkel
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- Bioinformatics Core Facility, Philipps University, Marburg, Germany
| | - Silke Reinartz
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Robert Grosse
- Institut for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs University, Freiburg, Germany
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
- Institute for Translational Proteomics, Philipps University, Marburg, Germany
| | - Rolf Müller
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- ✉ Corresponding author: Rolf Müller, Center for Tumor Biology and Immunology (ZTI), Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany. . Phone: +49 6421 2866236
| |
Collapse
|
35
|
Liu X, Lu Y, Huang J, Xing Y, Dai H, Zhu L, Li S, Feng J, Zhou B, Li J, Xia Q, Li J, Huang M, Gu Y, Su S. CD16 + fibroblasts foster a trastuzumab-refractory microenvironment that is reversed by VAV2 inhibition. Cancer Cell 2022; 40:1341-1357.e13. [PMID: 36379207 DOI: 10.1016/j.ccell.2022.10.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/16/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
The leukocyte Fcγ receptor (FcγR)-mediated response is important for the efficacy of therapeutic antibodies; however, little is known about the role of FcγRs in other cell types. Here we identify a subset of fibroblasts in human breast cancer that express CD16 (FcγRIII). An abundance of these cells in HER2+ breast cancer patients is associated with poor prognosis and response to trastuzumab. Functionally, upon trastuzumab stimulation, CD16+ fibroblasts reduce drug delivery by enhancing extracellular matrix stiffness. Interaction between trastuzumab and CD16 activates the intracellular SYK-VAV2-RhoA-ROCK-MLC2-MRTF-A pathway, leading to elevated contractile force and matrix production. Targeting of a Rho family guanine nucleotide exchange factor, VAV2, which is indispensable for the function of CD16 in fibroblasts rather than leukocytes, reverses desmoplasia provoked by CD16+ fibroblasts. Collectively, our study reveals a role for the fibroblast FcγR in drug resistance, and suggests that VAV2 is an attractive target to augment the effects of antibody treatments.
Collapse
Affiliation(s)
- Xinwei Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Breast Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Yiwen Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jingying Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Huiqi Dai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Liling Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shunrong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jingwei Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Boxuan Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiaqian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qidong Xia
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiang Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yuanting Gu
- Department of Breast Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China; Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Biotherapy Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
36
|
Fu Y, Zhang X, Liang X, Chen Y, Chen Z, Xiao Z. CapG promoted nasopharyngeal carcinoma cell motility involving Rho motility pathway independent of ROCK. World J Surg Oncol 2022; 20:347. [PMID: 36258216 PMCID: PMC9580211 DOI: 10.1186/s12957-022-02808-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/03/2022] [Indexed: 11/28/2022] Open
Abstract
Background Gelsolin-like capping actin protein (CapG) modulates actin dynamics and actin-based motility with a debatable role in tumorigenic progression. The motility-associated functions and potential molecular mechanisms of CapG in nasopharyngeal carcinoma (NPC) remain unclear. Methods CapG expression was detected by immunohistochemistry in a cohort of NPC tissue specimens and by Western blotting assay in a variety of NPC cell lines. Loss of function and gain of function of CapG in scratch wound-healing and transwell assays were performed. Inactivation of Rac1 and ROCK with the specific small molecular inhibitors was applied to evaluate CapG’s role in NPC cell motility. GTP-bound Rac1 and phosphorylated-myosin light chain 2 (p-MLC2) were measured in the ectopic CapG overexpressing cells. Finally, CapG-related gene set enrichment analysis was conducted to figure out the significant CapG-associated pathways in NPC. Results CapG disclosed increased level in the poorly differentiated NPC tissues and highly metastatic cells. Knockdown of CapG reduced NPC cell migration and invasion in vitro, while ectopic CapG overexpression showed the opposite effect. Ectopic overexpression of CapG compensated for the cell motility loss caused by simultaneous inactivation of ROCK and Rac1 or inactivation of ROCK alone. GTP-bound Rac1 weakened, and p-MLC2 increased in the CapG overexpressing cells. Bioinformatics analysis validated a positive correlation of CapG with Rho motility signaling, while Rac1 motility pathway showed no significant relationship. Conclusions The present findings highlight the contribution of CapG to NPC cell motility independent of ROCK and Rac1. CapG promotes NPC cell motility at least partly through MLC2 phosphorylation and contradicts with Rac1 activation. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-022-02808-7.
Collapse
Affiliation(s)
- Ying Fu
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, 451191, Henan, China
| | - Xujun Liang
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yongheng Chen
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhuchu Chen
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhefeng Xiao
- Department of Pathology, NHC Key Laboratory of Cancer Proteomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
37
|
Rotshenker S. Galectin-3 (MAC-2) controls phagocytosis and macropinocytosis through intracellular and extracellular mechanisms. Front Cell Neurosci 2022; 16:949079. [PMID: 36274989 PMCID: PMC9581057 DOI: 10.3389/fncel.2022.949079] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Galectin-3 (Gal-3; formally named MAC-2) is a β-galactoside-binding lectin. Various cell types produce Gal-3 under either normal conditions and/or pathological conditions. Gal-3 can be present in cells' nuclei and cytoplasm, secreted from producing cells, and associated with cells' plasma membranes. This review focuses on how Gal-3 controls phagocytosis and macropinocytosis. Intracellular and extracellular Gal-3 promotes the phagocytosis of phagocytic targets/cargo (e.g., tissue debris and apoptotic cells) in “professional phagocytes” (e.g., microglia and macrophages) and “non-professional phagocytes” (e.g., Schwann cells and astrocytes). Intracellularly, Gal-3 promotes phagocytosis by controlling the “eat me” signaling pathways that phagocytic receptors generate, directing the cytoskeleton to produce the mechanical forces that drive the structural changes on which phagocytosis depends, protrusion and then retraction of filopodia and lamellipodia as they, respectively, engulf and then internalize phagocytic targets. Extracellularly, Gal-3 promotes phagocytosis by functioning as an opsonin, linking phagocytic targets to phagocytic receptors, activating them to generate the “eat me” signaling pathways. Macropinocytosis is a non-selective endocytic mechanism that various cells use to internalize the bulk of extracellular fluid and included materials/cargo (e.g., dissolved nutrients, proteins, and pathogens). Extracellular and intracellular Gal-3 control macropinocytosis in some types of cancer. Phagocytosed and macropinocytosed targets/cargo that reach lysosomes for degradation may rupture lysosomal membranes. Damaged lysosomal membranes undergo either repair or removal by selective autophagy (i.e., lysophagy) that intracellular Gal-3 controls.
Collapse
|
38
|
Hino N, Matsuda K, Jikko Y, Maryu G, Sakai K, Imamura R, Tsukiji S, Aoki K, Terai K, Hirashima T, Trepat X, Matsuda M. A feedback loop between lamellipodial extension and HGF-ERK signaling specifies leader cells during collective cell migration. Dev Cell 2022; 57:2290-2304.e7. [PMID: 36174555 DOI: 10.1016/j.devcel.2022.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 07/11/2022] [Accepted: 09/07/2022] [Indexed: 11/03/2022]
Abstract
Upon the initiation of collective cell migration, the cells at the free edge are specified as leader cells; however, the mechanism underlying the leader cell specification remains elusive. Here, we show that lamellipodial extension after the release from mechanical confinement causes sustained extracellular signal-regulated kinase (ERK) activation and underlies the leader cell specification. Live-imaging of Madin-Darby canine kidney (MDCK) cells and mouse epidermis through the use of Förster resonance energy transfer (FRET)-based biosensors showed that leader cells exhibit sustained ERK activation in a hepatocyte growth factor (HGF)-dependent manner. Meanwhile, follower cells exhibit oscillatory ERK activation waves in an epidermal growth factor (EGF) signaling-dependent manner. Lamellipodial extension at the free edge increases the cellular sensitivity to HGF. The HGF-dependent ERK activation, in turn, promotes lamellipodial extension, thereby forming a positive feedback loop between cell extension and ERK activation and specifying the cells at the free edge as the leader cells. Our findings show that the integration of physical and biochemical cues underlies the leader cell specification during collective cell migration.
Collapse
Affiliation(s)
- Naoya Hino
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria.
| | - Kimiya Matsuda
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuya Jikko
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Gembu Maryu
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Katsuya Sakai
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan; WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Ryu Imamura
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan; WPI-Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Shinya Tsukiji
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan; Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Kazuhiro Aoki
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Kenta Terai
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tsuyoshi Hirashima
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Hakubi Center, Kyoto University, Kyoto, Japan; Japan Science and Technology Agency, Presto, Kawaguchi, Japan
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, Barcelona 08028, Spain; Faculty of Medicine, University of Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Michiyuki Matsuda
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
39
|
Cunha AF, Matias AF, Dias C, Oliveira MB, Araújo NAM, Mano JF. Cell Response in Free-Packed Granular Systems. ACS APPLIED MATERIALS & INTERFACES 2022; 14:40469-40480. [PMID: 36044384 PMCID: PMC9773234 DOI: 10.1021/acsami.1c24095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The study of the interactions of living adherent cells with mechanically stable (visco)elastic materials enables understanding and exploitation of physiological phenomena mediated by cell-extracellular communication. Insights into the interaction of cells and surrounding objects with different stability patterns upon cell contact might unveil biological responses to engineer innovative applications. Here, we hypothesize that the efficiency of cell attachment, spreading, and movement across a free-packed granular bed of microparticles depends on the microparticle diameter, raising the possibility of a necessary minimum traction force for the reinforcement of cell-particle bonds and long-term cell adhesion. The results suggest that microparticles with diameters of 14-20 μm are prone to cell-mediated mobility, holding the potential of inducing early cell detachment, while objects with diameters from 38 to 85 μm enable long-lasting cell adhesion and proliferation. An in silico hybrid particle-based model that addresses the time-dependent biological mechanisms of cell adhesion is proposed, providing inspiration for engineering platforms to address healthcare-related challenges.
Collapse
Affiliation(s)
- Ana F. Cunha
- Department
of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - André F.
V. Matias
- Centro
de Física Teórica e Computacional, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Departamento
de Física, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Cristóvão
S. Dias
- Centro
de Física Teórica e Computacional, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Departamento
de Física, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Mariana B. Oliveira
- Department
of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Nuno A. M. Araújo
- Centro
de Física Teórica e Computacional, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Departamento
de Física, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - João F. Mano
- Department
of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
40
|
Smooth Muscle Myosin Localizes at the Leading Edge and Regulates the Redistribution of Actin-regulatory Proteins during Migration. Cells 2022; 11:cells11152334. [PMID: 35954178 PMCID: PMC9367404 DOI: 10.3390/cells11152334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Airway smooth muscle cell migration plays an essential role in airway development, repair, and remodeling. Smooth muscle myosin II has been traditionally thought to localize in the cytoplasm solely and regulates cell migration by affecting stress fiber formation and focal adhesion assembly. In this study, we unexpectedly found that 20-kDa myosin light chain (MLC20) and myosin-11 (MYH11), important components of smooth muscle myosin, were present at the edge of lamellipodia. The knockdown of MLC20 or MYH11 attenuated the recruitment of c-Abl, cortactinProfilin-1 (Pfn-1), and Abi1 to the cell edge. Moreover, myosin light chain kinase (MLCK) colocalized with integrin β1 at the tip of protrusion. The inhibition of MLCK attenuated the recruitment of c-Abl, cortactin, Pfn-1, and Abi1 to the cell edge. Furthermore, MLCK localization at the leading edge was reduced by integrin β1 knockdown. Taken together, our results demonstrate that smooth muscle myosin localizes at the leading edge and orchestrates the recruitment of actin-regulatory proteins to the tip of lamellipodia. Mechanistically, integrin β1 recruits MLCK to the leading edge, which catalyzes MLC20 phosphorylation. Activated myosin regulates the recruitment of actin-regulatory proteins to the leading edge, and promotes lamellipodial formation and migration.
Collapse
|
41
|
Chen S, Guo F, Liu X, Xi J, Xue M, Guo Y, Wen J, Dong L, Chen Z. Roles of the RhoA-ROCK Signaling Pathway in the Endothelial H 2S Production and Vasodilation in Rat Cerebral Arteries. ACS OMEGA 2022; 7:18498-18508. [PMID: 35694456 PMCID: PMC9178624 DOI: 10.1021/acsomega.2c00996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
Cerebral endothelial H2S protects against cerebral ischemia-reperfusion injury through vasodilation, but its cerebral vasodilation mechanism and regulation of production are poorly understood. The RhoA-ROCK pathway plays important roles in vascular function. In this study, the roles of this pathway in the endothelial H2S production and vasodilation in rat cerebral arteries were investigated. Acetylcholine significantly increased H2S-generating enzyme cystathionine-γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST) protein expressions and H2S production in rat cerebrovascular endothelial cells (ECs), but the increases were markedly decreased by the M receptor blocker atropine or the CSE inhibitor dl-propargylglycine. Pretreatment with dl-propargylglycine or the 3-MST inhibitor l-aspartic acid markedly reduced the acetylcholine-increased H2S; CSE protein expression and H2S levels in the ECs were obviously attenuated by the RhoA agonist U46619 but increased by the RhoA inhibitor C3 transferase. U46619 also reduced 3-MST protein expression; Acetylcholine markedly inhibited RhoA protein expression and activity, but the inhibition was obviously reversed by atropine, dl-propargylglycine, and l-aspartic acid, respectively; Acetylcholine-induced endothelium-dependent vasodilation in rat cerebral basilar artery was significantly attenuated by pretreatment with dl-propargylglycine or l-aspartic acid or RhoA inhibitor CCG-1423 or ROCK inhibitor KD025, and was further decreased by co-pretreatment with dl-propargylglycine (or l-aspartic acid) and CCG-1423 (or KD025); NaHS significantly relaxed rat cerebral basilar artery vascular smooth muscle cells and inhibited ROCK1/2 activities, phosphorylated myosin light chain (MLC) protein expression, and KCl-increased [Ca2+]i, but these relaxation and inhibitions were markedly attenuated by pretreatment with C3 transferase or ROCK inhibitor Y27632. Our results demonstrated that endothelial H2S production is promoted by activation of the M receptor but inhibited by the RhoA-ROCK pathway in rat cerebral arteries; the endothelial H2S induces cerebral vasodilation by inhibiting this pathway to reduce phosphorylation of MLC and [Ca2+]i in vascular smooth muscle cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhiwu Chen
- . Tel: (+86)-0551-65161133. Fax: (+86)-0551-65161123
| |
Collapse
|
42
|
Leguay K, Decelle B, Elkholi IE, Bouvier M, Côté JF, Carréno S. Interphase microtubule disassembly is a signaling cue that drives cell rounding at mitotic entry. J Cell Biol 2022; 221:213183. [PMID: 35482006 DOI: 10.1083/jcb.202109065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/03/2022] [Accepted: 04/05/2022] [Indexed: 11/22/2022] Open
Abstract
At mitotic entry, reorganization of the actomyosin cortex prompts cells to round-up. Proteins of the ezrin, radixin, and moesin family (ERM) play essential roles in this process by linking actomyosin forces to the plasma membrane. Yet, the cell-cycle signal that activates ERMs at mitotic entry is unknown. By screening a compound library using newly developed biosensors, we discovered that drugs that disassemble microtubules promote ERM activation. We further demonstrated that disassembly of interphase microtubules at mitotic entry directs ERM activation and metaphase cell rounding through GEF-H1, a Rho-GEF inhibited by microtubule binding, RhoA, and its kinase effector SLK. We finally demonstrated that GEF-H1 and Ect2, another Rho-GEF previously identified to control actomyosin forces, act together to drive activation of ERMs and cell rounding in metaphase. In summary, we report microtubule disassembly as a cell-cycle signal that controls a signaling network ensuring that actomyosin forces are efficiently integrated at the plasma membrane to promote cell rounding at mitotic entry.
Collapse
Affiliation(s)
- Kévin Leguay
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada.,Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Université de Montréal, Montréal, Quebec, Canada
| | - Barbara Decelle
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada.,Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Université de Montréal, Montréal, Quebec, Canada
| | - Islam E Elkholi
- Montréal Clinical Research Institute, Montréal, Quebec, Canada.,Cytoskeletal Organization and Cell Migration lab, Université de Montréal, Montréal, Quebec, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada.,institution>Molecular Pharmacology Lab, Université de Montréal, Montréal, Quebec, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Jean-François Côté
- Montréal Clinical Research Institute, Montréal, Quebec, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Quebec, Canada.,Department of Medicine, McGill University, Montréal, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, Quebec, Canada.,Cytoskeletal Organization and Cell Migration lab, Université de Montréal, Montréal, Quebec, Canada
| | - Sébastien Carréno
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada.,Cellular Mechanisms of Morphogenesis during Mitosis and Cell Motility lab, Université de Montréal, Montréal, Quebec, Canada.,Department of Pathology and Cell Biology, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
43
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
44
|
Moonwiriyakit A, Pathomthongtaweechai N, Steinhagen PR, Chantawichitwong P, Satianrapapong W, Pongkorpsakol P. Tight junctions: from molecules to gastrointestinal diseases. Tissue Barriers 2022; 11:2077620. [PMID: 35621376 PMCID: PMC10161963 DOI: 10.1080/21688370.2022.2077620] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Intestinal epithelium functions as a tissue barrier to prevent interaction between the internal compartment and the external milieu. Intestinal barrier function also determines epithelial polarity for the absorption of nutrients and the secretion of waste products. These vital functions require strong integrity of tight junction proteins. In fact, intestinal tight junctions that seal the paracellular space can restrict mucosal-to-serosal transport of hostile luminal contents. Tight junctions can form both an absolute barrier and a paracellular ion channel. Although defective tight junctions potentially lead to compromised intestinal barrier and the development and progression of gastrointestinal (GI) diseases, no FDA-approved therapies that recover the epithelial tight junction barrier are currently available in clinical practice. Here, we discuss the impacts and regulatory mechanisms of tight junction disruption in the gut and related diseases. We also provide an overview of potential therapeutic targets to restore the epithelial tight junction barrier in the GI tract.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Nutthapoom Pathomthongtaweechai
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Peter R Steinhagen
- Department of Hepatology and Gastroenterology, Charité Medical School, Berlin, Germany
| | | | | | - Pawin Pongkorpsakol
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
45
|
Dede Eren A, Vermeulen S, Schmitz TC, Foolen J, de Boer J. The loop of phenotype: Dynamic reciprocity links tenocyte morphology to tendon tissue homeostasis. Acta Biomater 2022; 163:275-286. [PMID: 35584748 DOI: 10.1016/j.actbio.2022.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/24/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
Cells and their surrounding extracellular matrix (ECM) are engaged in dynamic reciprocity to maintain tissue homeostasis: cells deposit ECM, which in turn presents the signals that define cell identity. This loop of phenotype is obvious for biochemical signals, such as collagens, which are produced by and presented to cells, but the role of biomechanical signals is also increasingly recognised. In addition, cell shape goes hand in hand with cell function and tissue homeostasis. Aberrant cell shape and ECM is seen in pathological conditions, and control of cell shape in micro-fabricated platforms disclose the causal relationship between cell shape and cell function, often mediated by mechanotransduction. In this manuscript, we discuss the loop of phenotype for tendon tissue homeostasis. We describe cell shape and ECM organization in normal and diseased tissue, how ECM composition influences tenocyte shape, and how that leads to the activation of signal transduction pathways and ECM deposition. We further describe the use of technologies to control cell shape to elucidate the link between cell shape and its phenotypical markers and focus on the causal role of cell shape in the loop of phenotype. STATEMENT OF SIGNIFICANCE: The dynamic reciprocity between cells and their surrounding extracellular matrix (ECM) influences biomechanical and biochemical properties of ECM as well as cell function through activation of signal transduction pathways that regulate gene and protein expression. We refer to this reciprocity as Loop of Phenotype and it has been studied and demonstrated extensively by using micro-fabricated platforms to manipulate cell shape and cell fate. In this manuscript, we discuss this concept in tendon tissue homeostasis by giving examples in healthy and pathological tenson tissue. Furthermore, we elaborate this by showing how biomaterials are used to feed this reciprocity to manipulate cell shape and function. Finally, we elucidate the link between cell shape and its phenotypical markers and focus on the activation of signal transduction pathways and ECM deposition.
Collapse
Affiliation(s)
- Aysegul Dede Eren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Steven Vermeulen
- Maastricht University, MERLN Institute for Technology Inspired Regenerative Medicine, Instructive Biomaterial Engineering, Maastricht, the Netherlands
| | - Tara C Schmitz
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jasper Foolen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
46
|
Kawaguchi K, Asano S. Pathophysiological Roles of Actin-Binding Scaffold Protein, Ezrin. Int J Mol Sci 2022; 23:ijms23063246. [PMID: 35328667 PMCID: PMC8952289 DOI: 10.3390/ijms23063246] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023] Open
Abstract
Ezrin is one of the members of the ezrin/radixin/moesin (ERM) family of proteins. It was originally discovered as an actin-binding protein in the microvilli structure about forty years ago. Since then, it has been revealed as a key protein with functions in a variety of fields including cell migration, survival, and signal transduction, as well as functioning as a structural component. Ezrin acts as a cross-linker of membrane proteins or phospholipids in the plasma membrane and the actin cytoskeleton. It also functions as a platform for signaling molecules at the cell surface. Moreover, ezrin is regarded as an important target protein in cancer diagnosis and therapy because it is a key protein involved in cancer progression and metastasis, and its high expression is linked to poor survival in many cancers. Small molecule inhibitors of ezrin have been developed and investigated as candidate molecules that suppress cancer metastasis. Here, we wish to comprehensively review the roles of ezrin from the pathophysiological points of view.
Collapse
|
47
|
Tsukamoto S, Chiam KH, Asakawa T, Sawasaki K, Takesue N, Sakamoto N. Compressive forces driven by lateral actin fibers are a key to the nuclear deformation under uniaxial cell-substrate stretching. Biochem Biophys Res Commun 2022; 597:37-43. [PMID: 35123264 DOI: 10.1016/j.bbrc.2022.01.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 01/10/2023]
Abstract
Cells sense the direction of mechanical stimuli including substrate stretching and show morphological and functional responses. The nuclear deformation with respect to the direction of mechanical stimuli is thought of as a vital factor in mechanosensitive intracellular signaling and gene transcription, but the detailed relationship between the direction of stimuli and nuclear deformation behavior is not fully solved yet. Here, we assessed the role of actin cytoskeletons in nuclear deformation caused by cell substrate stretching with different directions. Cells on a PDMS stretching chamber were subjected to a step-strain and changes of long- and short-axes of nucleus before and after stretching were evaluated in terms of nuclear orientation against the direction of stretching. Nuclei oriented parallel to the stretching direction showed elongation and shrinkage in the long and short axes, respectively, and vice versa. However, calculation of the aspect ratio (ratio of long- and short-axes) changes revealed orientation-depend nuclear deformation: The nucleus oriented parallel to the stretching direction showed a greater aspect ratio change than it aligned in the perpendicular direction of the stretching. A decrease in actin cytoskeletal tension significantly changed the nuclear deformation only in the short axis direction, thereby abolishing the orientation-depend deformation of the nucleus. These results suggest that lateral compressive forces exerted by the actin cytoskeleton is a key factor of orientation-depend deformation in short axis of the nucleus under the cell-substrate stretching condition, and may be crucial for mechano-sensing and responses to the cell-substrate stretching direction.
Collapse
Affiliation(s)
- Shingo Tsukamoto
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan; Bioinformatics Institute, A∗STAR, Singapore.
| | | | - Takumi Asakawa
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Kaoru Sawasaki
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Naoyuki Takesue
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan.
| |
Collapse
|
48
|
Ali R, Mir HA, Hamid R, Bhat B, Shah RA, Khanday FA, Bhat SS. Actin Modulation Regulates the Alpha-1-Syntrophin/p66Shc Mediated Redox Signaling Contributing to the RhoA GTPase Protein Activation in Breast Cancer Cells. Front Oncol 2022; 12:841303. [PMID: 35273919 PMCID: PMC8904154 DOI: 10.3389/fonc.2022.841303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
SNTA1 signaling axis plays an essential role in cytoskeletal organization and is also implicated in breast cancers. In this study, we aimed to investigate the involvement of actin cytoskeleton in the propagation of SNTA1/p66shc mediated pro-metastatic cascade in breast cancer cells.The effect of actin filament depolymerization on SNTA1-p66Shc interaction and the trimeric complex formation was analyzed using co-immunoprecipitation assays. Immunofluorescence and RhoA activation assays were used to show the involvement of SNTA1-p66Shc interaction in RhoA activation and F-actin organization. Cellular proliferation and ROS levels were assessed using MTT assay and Amplex red catalase assay. The migratory potential was evaluated using transwell migration assay and wound healing assay.We found that cytochalasin D mediated actin depolymerization significantly declines endogenous interaction between SNTA1 and p66Shc protein in MDA-MB-231 cells. Results indicate that SNTA1 and p66Shc interact with RhoA protein under physiological conditions. The ROS generation and RhoA activation were substantially enhanced in cells overexpressing SNTA1 and p66Shc, promoting proliferation and migration in these cells. In addition, we found that loss of SNTA1-p66Shc interaction impaired actin organization, proliferation, and migration in breast cancer cells. Our results demonstrate a novel reciprocal regulatory mechanism between actin modulation and SNTA1/p66Shc/RhoA signaling cascade in human metastatic breast cancer cells.
Collapse
Affiliation(s)
- Roshia Ali
- Department of Biotechnology, University of Kashmir, Srinagar, India.,Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Hilal Ahmad Mir
- Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Rabia Hamid
- Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Basharat Bhat
- National Agricultural Higher Education Project (NAHEP) Sher-e-Kashmir University of Agricultural Sciences and Technology-Kashmir, Srinagar, India
| | - Riaz A Shah
- Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology-Kashmir, Faculty of Veterinary Sciences and Animal Husbandry, Srinagar, India
| | | | - Sahar Saleem Bhat
- Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology-Kashmir, Faculty of Veterinary Sciences and Animal Husbandry, Srinagar, India
| |
Collapse
|
49
|
Mohammadalipour A, Diaz MF, Livingston M, Ewere A, Zhou A, Horton PD, Olamigoke LT, Lamar JM, Hagan JP, Lee HJ, Wenzel PL. RhoA-ROCK competes with YAP to regulate amoeboid breast cancer cell migration in response to lymphatic-like flow. FASEB Bioadv 2022; 4:342-361. [PMID: 35520391 PMCID: PMC9065582 DOI: 10.1096/fba.2021-00055] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/16/2021] [Accepted: 01/26/2022] [Indexed: 11/11/2022] Open
Abstract
Lymphatic drainage generates force that induces prostate cancer cell motility via activation of Yes-associated protein (YAP), but whether this response to fluid force is conserved across cancer types is unclear. Here, we show that shear stress corresponding to fluid flow in the initial lymphatics modifies taxis in breast cancer, whereas some cell lines use rapid amoeboid migration behavior in response to fluid flow, a separate subset decrease movement. Positive responders displayed transcriptional profiles characteristic of an amoeboid cell state, which is typical of cells advancing at the edges of neoplastic tumors. Regulation of the HIPPO tumor suppressor pathway and YAP activity also differed between breast subsets and prostate cancer. Although subcellular localization of YAP to the nucleus positively correlated with overall velocity of locomotion, YAP gain- and loss-of-function demonstrates that YAP inhibits breast cancer motility but is outcompeted by other pro-taxis mediators in the context of flow. Specifically, we show that RhoA dictates response to flow. GTPase activity of RhoA, but not Rac1 or Cdc42 Rho family GTPases, is elevated in cells that positively respond to flow and is unchanged in cells that decelerate under flow. Disruption of RhoA or the RhoA effector, Rho-associated kinase (ROCK), blocked shear stress-induced motility. Collectively, these findings identify biomechanical force as a regulator amoeboid cell migration and demonstrate stratification of breast cancer subsets by flow-sensing mechanotransduction pathways.
Collapse
Affiliation(s)
- Amina Mohammadalipour
- Department of Integrative Biology & PharmacologyThe University of Texas Health Science Center at HoustonTexasUSA
| | - Miguel F. Diaz
- Department of Integrative Biology & PharmacologyThe University of Texas Health Science Center at HoustonTexasUSA,Children’s Regenerative Medicine ProgramDepartment of Pediatric SurgeryThe University of Texas Health Science Center at HoustonTexasUSA,Center for Stem Cell and Regenerative MedicineBrown Foundation Institute of Molecular MedicineThe University of Texas Health Science Center at HoustonTexasUSA
| | - Megan Livingston
- Department of Integrative Biology & PharmacologyThe University of Texas Health Science Center at HoustonTexasUSA,Children’s Regenerative Medicine ProgramDepartment of Pediatric SurgeryThe University of Texas Health Science Center at HoustonTexasUSA,Center for Stem Cell and Regenerative MedicineBrown Foundation Institute of Molecular MedicineThe University of Texas Health Science Center at HoustonTexasUSA,Biochemistry and Cell Biology ProgramMD Anderson UTHealth Graduate School of Biomedical SciencesThe University of TexasHoustonTexasUSA
| | - Adesuwa Ewere
- Children’s Regenerative Medicine ProgramDepartment of Pediatric SurgeryThe University of Texas Health Science Center at HoustonTexasUSA,Center for Stem Cell and Regenerative MedicineBrown Foundation Institute of Molecular MedicineThe University of Texas Health Science Center at HoustonTexasUSA,School of MedicineUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Allen Zhou
- Children’s Regenerative Medicine ProgramDepartment of Pediatric SurgeryThe University of Texas Health Science Center at HoustonTexasUSA,Center for Stem Cell and Regenerative MedicineBrown Foundation Institute of Molecular MedicineThe University of Texas Health Science Center at HoustonTexasUSA
| | - Paulina D. Horton
- Department of Integrative Biology & PharmacologyThe University of Texas Health Science Center at HoustonTexasUSA,Children’s Regenerative Medicine ProgramDepartment of Pediatric SurgeryThe University of Texas Health Science Center at HoustonTexasUSA,Center for Stem Cell and Regenerative MedicineBrown Foundation Institute of Molecular MedicineThe University of Texas Health Science Center at HoustonTexasUSA,Immunology ProgramMD Anderson UTHealth Graduate School of Biomedical SciencesThe University of TexasHoustonTexasUSA
| | - Loretta T. Olamigoke
- Vivian L. Smith Department of NeurosurgeryThe University of Texas Health Science Center at HoustonTexasUSA
| | - John M. Lamar
- Molecular and Cellular PhysiologyAlbany Medical CollegeAlbanyNew YorkUSA
| | - John P. Hagan
- Vivian L. Smith Department of NeurosurgeryThe University of Texas Health Science Center at HoustonTexasUSA
| | - Hyun J. Lee
- Department of Anatomy and Cell BiologyCollege of MedicineChung‐Ang UniversitySeoulSouth Korea,Department of Global Innovative DrugsGraduate School of Chung‐Ang UniversitySeoulSouth Korea
| | - Pamela L. Wenzel
- Department of Integrative Biology & PharmacologyThe University of Texas Health Science Center at HoustonTexasUSA,Children’s Regenerative Medicine ProgramDepartment of Pediatric SurgeryThe University of Texas Health Science Center at HoustonTexasUSA,Center for Stem Cell and Regenerative MedicineBrown Foundation Institute of Molecular MedicineThe University of Texas Health Science Center at HoustonTexasUSA,Biochemistry and Cell Biology ProgramMD Anderson UTHealth Graduate School of Biomedical SciencesThe University of TexasHoustonTexasUSA,Immunology ProgramMD Anderson UTHealth Graduate School of Biomedical SciencesThe University of TexasHoustonTexasUSA
| |
Collapse
|
50
|
Fujiya T, Asanuma K, Koike T, Okata T, Saito M, Asano N, Imatani A, Masamune A. Nitric oxide could promote development of Barrett's esophagus by S-nitrosylation-induced inhibition of Rho-ROCK signaling in esophageal fibroblasts. Am J Physiol Gastrointest Liver Physiol 2022; 322:G107-G116. [PMID: 34786954 DOI: 10.1152/ajpgi.00124.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Barrett's esophagus arises in the process of wound healing in distal esophageal epithelium damaged by gastroesophageal reflux disease. Differentiation of fibroblast into myofibroblasts, a smooth muscle cell-like phenotype and tissue contraction are crucial processes in wound healing. No study has evaluated mechanism by which luminal esophageal nitric oxide (NO) affect Rho-associated coiled coil-forming protein kinase (Rho-ROCK) signaling pathway, a key factor of tissue contraction, in stromal fibroblasts to develop Barrett's esophagus. Using esophageal fibroblasts, we performed collagen-based cell contraction assays and evaluated influence of Rho-ROCK signaling in the exposure to acidic bile salts and NOC-9, which is an NO donor. We found that enhanced cell contraction induced by acidic bile salts was inhibited by NO, accompanied by decrease in phosphorylated myosin light chain expression and stress fiber formation. NO directly S-nitrosylated GTP-RhoA and consequently blocked Rho-ROCK signaling. Moreover, exposure to NO and Y27632, a Rho-ROCK signaling inhibitor, decreased α-SMA expression and increased bone morphogenetic protein-4 (BMP4) expression and secretion. These findings could account for the increased expression of BMP4 in the columnar epithelial cells and stromal fibroblasts in human Barrett's esophagus. NO could impair wound contraction by blocking the Rho-ROCK signaling pathway and promote the development of Barrett's esophagus.NEW & NOTEWORTHY Barrett's esophagus is the condition where esophageal epithelium damaged by gastroesophageal reflux disease (GERD) is abnormally healed via replacing of metaplastic columnar epithelium, but very few studies have conducted focusing wound healing in the development of Barrett's esophagus. Esophageal luminal nitric oxide inhibits Rho-ROCK signaling pathway in esophageal fibroblasts, which leads to delay tissue contraction, a pivotal step in proper wound healing. Moreover, this inhibition increases tissue BMP4 expression. Impaired wound healing could be related to Barrett's esophagus.
Collapse
Affiliation(s)
- Taku Fujiya
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyotaka Asanuma
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoki Okata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiro Saito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Imatani
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|