1
|
Han X, Zhang C, Lei Q, Xu J, Zhou Y. Stiffness regulates extracellular matrix synthesis in fibroblasts by DDR1-TGF-β/STAT3 mechanotransduction axis. BIOMATERIALS ADVANCES 2025; 172:214240. [PMID: 40023083 DOI: 10.1016/j.bioadv.2025.214240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
For a long time, research on atherosclerosis (AS) has mainly focused on endothelial cells (ECs) and smooth muscle cells (SMCs) in blood vessels. Fibroblasts, however, being the major component in adventitia, little is known about their role. Fibroblasts are highly plastic cells, capable of undergoing phenotypic changes in response to various extracellular signals. Once activated, fibroblasts can promote fibrosis by altering the secretion of extracellular matrix (ECM). In this study, the effect of ECM stiffness on fibroblasts was investigated. Polyacrylamide (PA) gels with varying elastic moduli (1 kPa, 20 kPa and 100 kPa) were used as models for matrix stiffness. Human fibroblasts were cultured on these substrates, and their phenotypic and functional changes were examined. The data revealed that a collagen-binding receptor, Discoidin Domain Receptor 1 (DDR1), plays a central role in sensing mechanical stimuli from ECM. Matrix stiffness-induced phosphorylation of DDR1 suppresses the synthesis of ECM proteins in fibroblasts. The expression of ECM proteins on the 1 kPa substrate was significantly higher than that on the 20 kPa and 100 kPa substrates, while the phosphorylation level of DDR1 was notably reduced. After knocking out DDR1, the difference in ECM proteins expression among the three substrates with different stiffness levels disappeared. The signal transduction from DDR1 to ECM synthesis is mediated by the TGF-β/STAT3 signaling axis. Our study reveals how matrix stiffness regulates the synthesis of ECM in fibroblasts and paves the way for understanding the regulation of fibrotic process in the pathogenesis of AS.
Collapse
Affiliation(s)
- Xiaomei Han
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Chao Zhang
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Qian Lei
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Jin Xu
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Yue Zhou
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China.
| |
Collapse
|
2
|
Phillips M, Nimmo M, Rugonyi S. Developmental and Evolutionary Heart Adaptations Through Structure-Function Relationships. J Cardiovasc Dev Dis 2025; 12:83. [PMID: 40137081 PMCID: PMC11942974 DOI: 10.3390/jcdd12030083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
While the heart works as an efficient pump, it also has a high level of adaptivity by changing its structure to maintain function during healthy and diseased states. In this Review, we present examples of structure-function relationships across species and throughout embryonic development in mammals and birds. We also summarize current research on avian models aiming at understanding how biophysical and biological mechanisms closely interact during heart formation. We conclude by underscoring similarities between cardiac adaptations and structural changes over developmental and evolutionary time scales and how understanding the mechanisms behind these adaptations can help prevent or alleviate the effects of cardiac malformations and contribute to cardiac regeneration efforts.
Collapse
Affiliation(s)
| | | | - Sandra Rugonyi
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97239, USA; (M.P.); (M.N.)
| |
Collapse
|
3
|
Oak ASW, Bagchi A, Brukman MJ, Toth J, Ford J, Zheng Y, Nace A, Yang R, Hsieh JC, Hayden JE, Ruthel G, Ray A, Kim E, Shenoy VB, Cotsarelis G. Wnt signaling modulates mechanotransduction in the epidermis to drive hair follicle regeneration. SCIENCE ADVANCES 2025; 11:eadq0638. [PMID: 39970220 PMCID: PMC11838001 DOI: 10.1126/sciadv.adq0638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Most wounds form scars without hair follicles. However, in the wound-induced hair neogenesis (WIHN) model of skin regeneration, wounds regenerate hair follicles if tissue rigidity is optimal. Although WIHN depends on Wnt signaling, whether Wnt performs a mechanoregulatory role that contributes to regeneration remains uncharacterized. Here, we demonstrate that Wnt signaling affects mechanosensitivity at both cellular and tissue levels to drive WIHN. Atomic force microscopy revealed an attenuated substrate rigidity response in epidermal but not dermal cells of healing wounds. Super-resolution microscopy and nanoneedle probing of intracellular compartments in live human keratinocytes revealed that Wnt-induced chromatin remodeling triggers a 10-fold drop in nuclear rigidity without jeopardizing the nucleocytoskeletal mechanical coupling. Mechanistically, Wnt signaling orchestrated a massive reorganization of actin architecture and recruited adherens junctions to generate a mechanical syncytium-a cohesive contractile unit with superior capacity for force coordination and collective durotaxis. Collectively, our findings unveil Wnt signaling's mechanoregulatory role that manipulates the machinery of mechanotransduction to drive regeneration.
Collapse
Affiliation(s)
- Allen S. W. Oak
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amrit Bagchi
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew J. Brukman
- Singh Center for Nanotechnology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua Toth
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jamie Ford
- Singh Center for Nanotechnology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Zheng
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arben Nace
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruifeng Yang
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jen-Chih Hsieh
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anisa Ray
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine Kim
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vivek B. Shenoy
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - George Cotsarelis
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Mascharak S, Griffin M, Talbott HE, Guo JL, Parker J, Morgan AG, Valencia C, Kuhnert MM, Li DJ, Liang NE, Kratofil RM, Daccache JA, Sidhu I, Davitt MF, Guardino N, Lu JM, Abbas DB, Deleon NMD, Lavin CV, Adem S, Khan A, Chen K, Henn D, Spielman A, Cotterell A, Akras D, Downer M, Tevlin R, Lorenz HP, Gurtner GC, Januszyk M, Naik S, Wan DC, Longaker MT. Inhibiting mechanotransduction prevents scarring and yields regeneration in a large animal model. Sci Transl Med 2025; 17:eadt6387. [PMID: 39970235 DOI: 10.1126/scitranslmed.adt6387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Modulating mechanotransduction by inhibiting yes-associated protein (YAP) in mice yields wound regeneration without scarring. However, rodents are loose-skinned and fail to recapitulate key aspects of human wound repair. We sought to elucidate the effects of YAP inhibition in red Duroc pig wounds, the most human-like model of scarring. We show that one-time treatment with verteporfin, a YAP inhibitor, immediately after wounding is sufficient to prevent scarring and to drive wound regeneration in pigs. By performing single-cell RNA sequencing (scRNA-seq) on porcine wounds in conjunction with spatial proteomic analysis, we found perturbations in fibroblast dynamics with verteporfin treatment and the presence of putative pro-regenerative/profibrotic fibroblasts enriched in regenerating/scarring pig wounds, respectively. We also identified differences in enriched myeloid cell subpopulations after treatment and linked this observation to increased elaboration of interleukin-33 (IL-33) in regenerating wounds. Finally, we validated our findings in a xenograft wound model containing human neonatal foreskin engrafted onto nude mice and used scRNA-seq of human wound cells to draw parallels with fibroblast subpopulation dynamics in porcine wounds. Collectively, our findings provide support for the clinical translation of local mechanotransduction inhibitors to prevent human skin scarring, and they clarify a YAP/IL-33 signaling axis in large animal wound regeneration.
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heather E Talbott
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jason L Guo
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer Parker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Annah Grace Morgan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caleb Valencia
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maxwell Michael Kuhnert
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dayan J Li
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Norah E Liang
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel M Kratofil
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joseph A Daccache
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ikjot Sidhu
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, NY 10016, USA
| | - Michael F Davitt
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicholas Guardino
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John M Lu
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Darren B Abbas
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nestor M D Deleon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christopher V Lavin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sandeep Adem
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anum Khan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kellen Chen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dominic Henn
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amanda Spielman
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Asha Cotterell
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deena Akras
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mauricio Downer
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ruth Tevlin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Peter Lorenz
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Geoffrey C Gurtner
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shruti Naik
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald O. Perelman Department of Dermatology, NYU Langone Health, New York, NY 10016, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
5
|
Hoffman CL, Bharathan NK, Shibata Y, Giang W, Gudjonsson JE, Seykora JT, Prouty SM, Stahley SN, Payne AS, Kowalczyk AP. Pemphigus Vulgaris Autoantibodies Induce an Endoplasmic Reticulum Stress Response. J Invest Dermatol 2025:S0022-202X(25)00085-5. [PMID: 39909113 DOI: 10.1016/j.jid.2024.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/12/2024] [Accepted: 12/26/2024] [Indexed: 02/07/2025]
Abstract
Desmosomes are intercellular junctions that mediate cell-cell adhesion and are essential for maintaining tissue integrity. Pemphigus vulgaris (PV) is an autoimmune epidermal blistering disease caused by autoantibodies (IgG) targeting desmoglein 3, a desmosomal cadherin. PV autoantibodies cause desmosome disassembly and loss of cell-cell adhesion; however, the molecular signaling pathways that regulate these processes are not fully understood. Using high-resolution time-lapse imaging of live keratinocytes, we found that endoplasmic reticulum (ER) tubules make frequent and persistent contacts with internalizing desmoglein 3 puncta in keratinocytes treated with IgG of patients with PV. Biochemical experiments demonstrated that PV IgG activated ER stress signaling pathways, including both IRE1⍺ and PERK pathways, in cultured keratinocytes. Furthermore, ER stress transcripts were upregulated in the skin of patients with PV. Pharmacological inhibition of ER stress protects against PV IgG-induced desmosome disruption and loss of keratinocyte cell-cell adhesion, suggesting that ER stress may be an important pathomechanism and a therapeutically targetable pathway for PV treatment. These data support a model in which desmosome adhesion is integrated with ER function to serve as a cell adhesion stress sensor that is activated in blistering skin diseases.
Collapse
Affiliation(s)
- Coryn L Hoffman
- Department of Dermatology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | | | - Yoshitaka Shibata
- Department of Dermatology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - William Giang
- Department of Dermatology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - John T Seykora
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen M Prouty
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sara N Stahley
- Department of Dermatology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Aimee S Payne
- Department of Dermatology, Columbia University, New York, New York, USA
| | - Andrew P Kowalczyk
- Department of Dermatology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
6
|
Le PM, Quinn JM, Flozak AS, Steffeck AWT, Huang CF, Gottardi CJ. α-catenin phosphorylation is elevated during mitosis to resist apical rounding and epithelial barrier leak. Biol Open 2025; 14:bio061726. [PMID: 39782767 PMCID: PMC11744050 DOI: 10.1242/bio.061726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
Epithelial cell cohesion and barrier function critically depend on α-catenin, an actin-binding protein and essential constituent of cadherin-catenin-based adherens junctions. α-catenin undergoes actomyosin force-dependent unfolding of both actin-binding and middle domains to strongly engage actin filaments and its various effectors; this mechanosensitivity is critical for adherens junction function. We previously showed that α-catenin is highly phosphorylated in an unstructured region that links the mechanosensitive middle and actin-binding domains (known as the P-linker region), but the cellular processes that promote α-catenin phosphorylation have remained elusive. Here, we leverage a previously published phospho-proteomic data set to show that the α-catenin P-linker region is maximally phosphorylated during mitosis. By reconstituting α-catenin CRISPR knockout MDCK cells with wild-type, phospho-mutant and phospho-mimic forms of α-catenin, we show that full phosphorylation restrains mitotic cell rounding in the apical direction, strengthening the interactions between dividing and non-dividing neighbors to limit epithelial barrier leak. As the major scaffold components of adherens junctions, tight junctions and desmosomes are also differentially phosphorylated during mitosis, we reason that epithelial cell division may be a tractable system to understand how junction complexes are coordinately regulated to sustain barrier function under tension-generating morphogenetic processes.
Collapse
Affiliation(s)
- Phuong M. Le
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeanne M. Quinn
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Driskill Graduate Program in the Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Annette S. Flozak
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Adam W. T. Steffeck
- Driskill Graduate Program in the Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Che-Fan Huang
- Proteomics Center of Excellence, Northwestern University, Evanston, IL 60208, USA
| | - Cara J. Gottardi
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Cell & Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
7
|
Outla Z, Prechova M, Korelova K, Gemperle J, Gregor M. Mechanics of cell sheets: plectin as an integrator of cytoskeletal networks. Open Biol 2025; 15:240208. [PMID: 39875099 PMCID: PMC11774597 DOI: 10.1098/rsob.240208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Epithelia are multicellular sheets that form barriers defining the internal and external environments. The constant stresses acting at this interface require that epithelial sheets are mechanically robust and provide a selective barrier to the hostile exterior. These properties are mediated by cellular junctions which are physically linked with heavily crosslinked cytoskeletal networks. Such hardwiring is facilitated by plakins, a family of giant modular proteins which serve as 'molecular bridges' between different cytoskeletal filaments and multiprotein adhesion complexes. Dysfunction of cytoskeletal crosslinking compromises epithelial biomechanics and structural integrity. Subsequent loss of barrier function leads to disturbed tissue homeostasis and pathological consequences such as skin blistering or intestinal inflammation. In this article, we highlight the importance of the cytolinker protein plectin for the functional organization of epithelial cytoskeletal networks. In particular, we focus on the ability of plectin to act as an integrator of the epithelial cytoarchitecture that defines the biomechanics of the whole tissue. Finally, we also discuss the role of cytoskeletal crosslinking in emerging aspects of epithelial mechanobiology that are critical for the maintenance of epithelial homeostasis.
Collapse
Affiliation(s)
- Zuzana Outla
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Magdalena Prechova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Katerina Korelova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Gemperle
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
8
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Santio NM, Ganesh K, Kaipainen PP, Halme A, Seyednasrollah F, Arbash E, Hänninen S, Kivelä R, Carpen O, Saharinen P. Endothelial Pim3 kinase protects the vascular barrier during lung metastasis. Nat Commun 2024; 15:10514. [PMID: 39627185 PMCID: PMC11615401 DOI: 10.1038/s41467-024-54445-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/07/2024] [Indexed: 12/06/2024] Open
Abstract
Endothelial cells (ECs) form a tissue-specific barrier for disseminating cancer cells in distant organs. However, the molecular regulation of the ECs in the metastatic niche remains unclear. Here, we analyze using scRNA-Seq, the transcriptional reprogramming of lung ECs six hours after the arrival of melanoma cells in mouse lungs. We discover a reactive capillary EC cluster (rCap) that increases from general capillary ECs in response to infiltrating cancer cells. rCap is enriched for angiogenic and inflammatory pathways and is also found in human lung datasets. The JAK-STAT activated oncogenic Pim3 kinase is a marker of rCap, being upregulated in spontaneous metastasis models. Notably, PIM inhibition increases vascular leakage and metastatic colonization and impairs the EC barrier by decreasing the junctional cadherin-5 and catenins α, β and δ. These results highlight the pulmonary endothelium's plasticity and its protection by PIM3, which may impair the efficacy of PIM inhibitors in cancer therapies.
Collapse
Affiliation(s)
- Niina M Santio
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Keerthana Ganesh
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Pihla P Kaipainen
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Aleksi Halme
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Fatemeh Seyednasrollah
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Emad Arbash
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Satu Hänninen
- Systems Oncology, Research Programs Unit University of Helsinki, Finland, Helsinki
| | - Riikka Kivelä
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Faculty of Sport and Health Sciences University of Jyväskylä, Jyväskylä, Finland
| | - Olli Carpen
- Systems Oncology, Research Programs Unit University of Helsinki, Finland, Helsinki
- Pathology/HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Pipsa Saharinen
- Translational Cancer Medicine, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland.
- Department of Biochemistry and Developmental Biology, Faculty of Medicine University of Helsinki, Helsinki, Finland.
| |
Collapse
|
10
|
Huveneers S, Phng LK. Endothelial cell mechanics and dynamics in angiogenesis. Curr Opin Cell Biol 2024; 91:102441. [PMID: 39342870 DOI: 10.1016/j.ceb.2024.102441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024]
Abstract
The efficient distribution of oxygen and metabolites is critical for embryonic development and growth as well as tissue homeostasis. This is achieved by endothelial cells forming and maintaining a closed, circulatory network of tubular blood vessels. Endothelial cells are highly plastic cells with the capability to generate diverse dynamic responses at different stages of vessel development in order to build vessel networks of tissue-specific patterns and morphologies. In this review, we discuss new conceptual advances gained from in vitro and in vivo models of angiogenesis on the control of endothelial cell dynamics. We highlight the complex interplay between mechanical cues, actin cytoskeleton and endothelial behaviors, and the emerging importance of hydrostatic pressure in complementing actin-dependent mechanisms to regulate endothelial cell mechanics and angiogenesis. Understanding these processes provides insights into vascular repair and regeneration mechanisms.
Collapse
Affiliation(s)
- Stephan Huveneers
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| | - Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
| |
Collapse
|
11
|
Wang X, Yao DS, Xu L, Yan DM, Zhao Y, Peng JH, Fu QL, Hu YY, Feng Q. Jianpi Huoxue Decoction Ameliorates Alcohol-associated Liver Disease by Improving Intestinal Barrier Function in Rats. Curr Med Sci 2024; 44:1241-1248. [PMID: 39617866 DOI: 10.1007/s11596-024-2955-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/08/2024] [Indexed: 12/24/2024]
Abstract
OBJECTIVE Jianpi huoxue decoction (JHD), a Chinese herbal formula, is commonly used for treating alcohol-associated liver disease (ALD). This study aimed to investigate the mechanism by which JHD affects intestinal barrier function in ALD rats. METHODS The Sprague-Dawley rats were randomly divided into three groups: control group, model group and JHD group. They were pair-fed a modified Lieber-DeCarli liquid diet containing alcohol (model group, n=10; JHD group, n=10) or isocaloric maltose dextrin (control group, n=10) for 6 weeks. After 3 weeks of feeding, the mice in the JHD group were given JHD (10 mL/kg/day) by gavage for 3 weeks, and those in the control and model groups received equal amounts of double-distilled water for the same period of time. Afterwards, all the rats were given lipopolysaccharide (LPS) by gavage and sacrificed 3.5 h later. LPS levels were measured in the portal blood to evaluate gut leakage. Transmission electron microscopy (TEM) was used to observe ultrastructural changes in the intestinal tract. Adherens junction (AJ) and tight junction (TJ) proteins were detected by Western blotting, immunofluorescence or immunohistochemistry. RESULTS JHD ameliorated Lieber-DeCarli liquid diet-induced hepatic steatosis, inflammation and LPS expression. It improved pathological changes in the liver and alleviated intestinal ultrastructure injury. Moreover, it significantly enhanced the integrity of tight junctions by increasing the expression of zonula occludens-1 (ZO-1) and occludin. It suppressed the activation of myosin light chain (MLC) phosphorylation. CONCLUSION JHD improves intestinal barrier function and reduces gut leakiness in ALD rats.
Collapse
Affiliation(s)
- Xin Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong-Sheng Yao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lin Xu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong-Ming Yan
- Clinical Pharmacokinetics Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu Zhao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing-Hua Peng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qi-Lin Fu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yi-Yang Hu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Clinical Pharmacokinetics Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
12
|
Nong Y, Lu J, Yu D, Wei X. Neohesperidin Dihydrochalcone Alleviates Lipopolysaccharide-Induced Vascular Endothelium Dysfunction by Regulating Antioxidant Capacity. Immun Inflamm Dis 2024; 12:e70107. [PMID: 39699295 PMCID: PMC11656606 DOI: 10.1002/iid3.70107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Endothelial dysfunction is one of the important mechanisms of organ and tissue damage in sepsis. In this study, we evaluated the effects of neohesperidin dihydrochalone (NHDC) on lipopolysaccharide (LPS)-induced vascular dysfunction and explored the potential mechanisms. METHODS In vivo, we assessed vascular leakage in mice by injecting Evans blue dye. In vitro, cell counting kit-8 (CCK-8) assay and flow cytometry were used to assess the activity of HUVEC and apoptosis. The effect of LPS on HUVEC barrier was assessed using FITC-extend membrane assay. The adhesion ability of HUVEC was tested by THP-1 cell adhesion assay. The antioxidant capacity of cells was measured by detecting the level of mitochondrial membrane potential, ROS, and content of CAT, SOD, GSH, and MDA within the cells. Furthermore, the release of endothelial IL-1β, IL-6, and TNF-α were detected by ELISA, and the expression level of TAK1, ERK1/2, and NFκB were detected by western blot. RESULTS Treatment with NHDC effectively alleviated LPS-induced endothelial permeability and organ damage by reducing reactive oxygen species production and enhancing the antioxidant response. Further investigation suggested that NHDC may exert its protective effects by inhibiting the release of IL-1β, IL-6, and TNF-α, and by decreasing the phosphorylation of key inflammatory signaling molecules, including transforming growth factor-β-activated kinase 1 (TAK1), extracellular signal-regulated kinases 1/2 (ERK1/2), and nuclear factor kappa B (NFκB). CONCLUSIONS Our study indicate that pretreatment with NHDC may provide protection against LPS-induced vascular dysfunction by reducing oxidative stress and activation of inflammatory signaling pathways.
Collapse
Affiliation(s)
- Yuxin Nong
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Junquan Lu
- Shantou University Medical CollegeShantouChina
| | - Danqing Yu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Xuebiao Wei
- Department of Geriatric Intensive Medicine, Guangdong Provincial Geriatrics InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouChina
| |
Collapse
|
13
|
Klein C, Ramminger I, Bai S, Steinberg T, Tomakidi P. Impairment of Intermediate Filament Expression Reveals Impact on Cell Functions Independent from Keratinocyte Transformation. Cells 2024; 13:1960. [PMID: 39682709 PMCID: PMC11640723 DOI: 10.3390/cells13231960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Although cytoplasmic intermediate filaments (cIFs) are essential for cell physiology, the molecular and cell functional consequences of cIF disturbances are poorly understood. Identifying defaults in cell function-controlled tissue homeostasis and understanding the interrelationship between specific cIFs and distinct cell functions remain key challenges. Using an RNAi-based mechanistic approach, we connected the impairment of cell-inherent cIFs with molecular and cell functional consequences, such as proliferation and differentiation. To investigate cIF disruption consequences in the oral epithelium, different cell transformation stages, originating from alcohol-treated oral gingival keratinocytes, were used. We found that impairment of keratin (KRT) KRT5, KRT14 and vimentin (VIM) affects proliferation and differentiation, and modulates the chromatin status. Furthermore, cIF impairment reduces the expression of nuclear integrity participant lamin B1 and the terminal keratinocyte differentiation marker involucrin (IVL). Conversely, impairment of IVL reduces cIF expression levels, functionally suggesting a regulatory interaction between cIFs and IVL. The findings demonstrate that the impairment of cIFs leads to imbalances in proliferation and differentiation, both of which are essential for tissue homeostasis. Thus, targeted impairment of cIFs appears promising to investigate the functional role of cIFs on cell-dependent tissue physiology at the molecular level and identifies putative interactions of cIFs with epithelial differentiation.
Collapse
Affiliation(s)
- Charlotte Klein
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (C.K.); (I.R.); (S.B.); (P.T.)
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Imke Ramminger
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (C.K.); (I.R.); (S.B.); (P.T.)
- Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104 Freiburg, Germany
| | - Shuoqiu Bai
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (C.K.); (I.R.); (S.B.); (P.T.)
| | - Thorsten Steinberg
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (C.K.); (I.R.); (S.B.); (P.T.)
| | - Pascal Tomakidi
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstr. 55, 79106 Freiburg, Germany; (C.K.); (I.R.); (S.B.); (P.T.)
| |
Collapse
|
14
|
Liu X, Min Q, Cheng X, Zhang W, Wu Q, Chen X, Lv M, Liu S, Zhao H, Yang D, Tai Y, Lei X, Wang Y, Zhan Q. Quiescent cancer cells induced by high-density cultivation reveals cholesterol-mediated survival and lung metastatic traits. Br J Cancer 2024; 131:1591-1604. [PMID: 39390252 PMCID: PMC11555385 DOI: 10.1038/s41416-024-02861-x] [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: 05/17/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND The metastatic cascade, a multifaceted and highly aggressive process, is the primary cause of mortality. The survival of quiescent cancer cells in circulatory system during metastasis is crucial, yet our comprehension is constrained by the absence of universally accepted quiescent cancer models. METHOD We developed a quiescent cancer cell model using high-density cultivation. Based on the scRNA-seq analysis, IP-MS, metabolomics, mouse lung metastasis models, cholesterol assay, PLA and other molecular experiments, we explored the molecular mechanism. Immunofluorescence, atomic force microscope, FluidFM, and shear stress stimulation were used to analyze the cytoskeleton and membrane properties contributing to mechanical force resistance. RESULT We established a quiescent cancer cell model induced by high-density cultivation. Single-cell RNA sequencing (scRNA-seq) analysis reveals that CDC25A plays a crucial role in the transition to quiescence, with its expression significantly elevated in the quiescent state. Depletion of CDC25A leads to an increased proliferative capacity, and reduced metastasis under high-density conditions. Mechanistically, upregulated CDC25A in quiescent cells enhances cholesterol metabolism via endosome pathways, leading to cell cycle arrest. This increase in cholesterol reinforces the cytoskeleton, alters membrane properties, and improves resistance to mechanical forces in circulatory system. CONCLUSION CDC25A significantly increased the cholesterol metabolism through endosome pathway in quiescent cancer cells, leading to the significant changes in cytoskeleton and membrane properties so as to enhance the resistance of mechanical force in circulatory system, facilitating lung metastasis. In high-density cultivation, quiescent cancer cells, up-regulate cholesterol metabolism by CDC25A through endosome pathway, enhancing the resistance to mechanical force in circulatory system, facilitating lung metastasis.
Collapse
Affiliation(s)
- Xingyang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Qinjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xinxin Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xu Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Mengzhu Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Siqi Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Huihui Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Di Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Yidi Tai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiao Lei
- Peking University International Cancer Institute, 100191, Beijing, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Peking University International Cancer Institute, 100191, Beijing, China.
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, 100730, Beijing, China.
- Soochow University Cancer Institute, Suzhou, 215000, China.
| |
Collapse
|
15
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
16
|
Janssen V, Huveneers S. Cell-cell junctions in focus - imaging junctional architectures and dynamics at high resolution. J Cell Sci 2024; 137:jcs262041. [PMID: 39480660 DOI: 10.1242/jcs.262041] [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] [Indexed: 11/02/2024] Open
Abstract
Studies utilizing electron microscopy and live fluorescence microscopy have significantly enhanced our understanding of the molecular mechanisms that regulate junctional dynamics during homeostasis, development and disease. To fully grasp the enormous complexity of cell-cell adhesions, it is crucial to study the nanoscale architectures of tight junctions, adherens junctions and desmosomes. It is important to integrate these junctional architectures with the membrane morphology and cellular topography in which the junctions are embedded. In this Review, we explore new insights from studies using super-resolution and volume electron microscopy into the nanoscale organization of these junctional complexes as well as the roles of the junction-associated cytoskeleton, neighboring organelles and the plasma membrane. Furthermore, we provide an overview of junction- and cytoskeletal-related biosensors and optogenetic probes that have contributed to these advances and discuss how these microscopy tools enhance our understanding of junctional dynamics across cellular environments.
Collapse
Affiliation(s)
- Vera Janssen
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Nosrati H, Fallah Tafti M, Aghamollaei H, Bonakdar S, Moosazadeh Moghaddam M. Directed Differentiation of Adipose-Derived Stem Cells Using Imprinted Cell-Like Topographies as a Growth Factor-Free Approach. Stem Cell Rev Rep 2024; 20:1752-1781. [PMID: 39066936 DOI: 10.1007/s12015-024-10767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
The influence of surface topography on stem cell behavior and differentiation has garnered significant attention in regenerative medicine and tissue engineering. The cell-imprinting method has been introduced as a promising approach to mimic the geometry and topography of cells. The cell-imprinted substrates are designed to replicate the topographies and dimensions of target cells, enabling tailored interactions that promote the differentiation of stem cells towards desired specialized cell types. In fact, by replicating the size and shape of cells, biomimetic substrates provide physical cues that profoundly impact stem cell differentiation. These cues play a pivotal role in directing cell morphology, cytoskeletal organization, and gene expression, ultimately influencing lineage commitment. The biomimetic substrates' ability to emulate the native cellular microenvironment supports the creation of platforms capable of steering stem cell fate with high precision. This review discusses the role of mechanical factors that impact stem cell fate. It also provides an overview of the design and fabrication principles of cell-imprinted substrates. Furthermore, the paper delves into the use of cell-imprinted polydimethylsiloxane (PDMS) substrates to direct adipose-derived stem cells (ADSCs) differentiation into a variety of specialized cells for tissue engineering and regenerative medicine applications. Additionally, the review discusses the limitations of cell-imprinted PDMS substrates and highlights the efforts made to overcome these limitations.
Collapse
Affiliation(s)
- Hamed Nosrati
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahsa Fallah Tafti
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran.
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Yaba A, Thalheim T, Schneider MR. The role of cell-cell and cell-matrix junctional complexes in sebaceous gland homeostasis and differentiation. Cell Commun Signal 2024; 22:445. [PMID: 39313816 PMCID: PMC11421122 DOI: 10.1186/s12964-024-01835-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024] Open
Abstract
Sebaceous glands (SG) are essential for maintaining skin integrity, as their lipid-rich secretion (sebum) lubricates and protects the epidermis and hairs. In addition, these glands have an emerging role in immunomodulation and may affect whole-body energy metabolism, besides being an appealing model for research in topics as lipogenesis, stem cell biology and tumorigenesis. In spite of the increasing interest in studying SGs pathophysiology, sebocyte cell-cell and cell-matrix adhesion processes have been only superficially examined, and never in a systematic way. This is regrettable considering the key role of cellular adhesion in general, the specific expression pattern of indivdual junctional complexes, and the reports of structural changes in SGs after altered expression of adhesion-relevant proteins. Here, we review the available information on structural and functional aspects of cell-cell and cell-matrix junctions in sebocytes, and how these processes change under pathological conditions. This information will contribute for better understanding sebocyte differentiation and sebum secretion, and may provide hints for novel therapeutic strategies for skin diseases.
Collapse
Affiliation(s)
- Aylin Yaba
- Department of Histology and Embryology, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Torsten Thalheim
- Present Address: Deutsches Biomasseforschungszentrum gGmbH, Torgauer Str. 116, 04347, Leipzig, Germany
- Interdisciplinary Centre for Bioinformatics, Härtelstr. 16-18, 04107, Leipzig, Germany
| | - Marlon R Schneider
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany.
| |
Collapse
|
19
|
Anney P, Charpentier P, Proulx S. Influence of Intraocular Pressure on the Expression and Activity of Sodium-Potassium Pumps in the Corneal Endothelium. Int J Mol Sci 2024; 25:10227. [PMID: 39337712 PMCID: PMC11432950 DOI: 10.3390/ijms251810227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
The corneal endothelium is responsible for pumping fluid out of the stroma in order to maintain corneal transparency, which depends in part on the expression and activity of sodium-potassium pumps. In this study, we evaluated how physiologic pressure and flow influence transcription, protein expression, and activity of Na+/K+-ATPase. Native and engineered corneal endothelia were cultured in a bioreactor in the presence of pressure and flow (hydrodynamic culture condition) or in a Petri dish (static culture condition). Transcription of ATP1A1 was assessed using qPCR, the expression of the α1 subunit of Na+/K+-ATPase was measured using Western blots and ELISA assays, and Na+/K+-ATPase activity was evaluated using an ATPase assay in the presence of ouabain. Results show that physiologic pressure and flow increase the transcription and the protein expression of Na+/K+-ATPase α1 in engineered corneal endothelia, while they remain stable in native corneal endothelia. Interestingly, the activity of Na+/K+-ATPase was increased in the presence of physiologic pressure and flow in both native and engineered corneal endothelia. These findings highlight the role of the in vivo environment on the functionality of the corneal endothelium.
Collapse
Affiliation(s)
- Princia Anney
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec City, QC G1J 1Z4, Canada
- Département d'Ophtalmologie et ORL-Chirurgie Cervico-Faciale, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Pascale Charpentier
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec City, QC G1J 1Z4, Canada
- Département d'Ophtalmologie et ORL-Chirurgie Cervico-Faciale, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Stéphanie Proulx
- Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC G1S 4L8, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec City, QC G1J 1Z4, Canada
- Département d'Ophtalmologie et ORL-Chirurgie Cervico-Faciale, Université Laval, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
20
|
Al-Nuaimi DA, Rütsche D, Abukar A, Hiebert P, Zanetti D, Cesarovic N, Falk V, Werner S, Mazza E, Giampietro C. Hydrostatic pressure drives sprouting angiogenesis via adherens junction remodelling and YAP signalling. Commun Biol 2024; 7:940. [PMID: 39097636 PMCID: PMC11297954 DOI: 10.1038/s42003-024-06604-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 07/17/2024] [Indexed: 08/05/2024] Open
Abstract
Endothelial cell physiology is governed by its unique microenvironment at the interface between blood and tissue. A major contributor to the endothelial biophysical environment is blood hydrostatic pressure, which in mechanical terms applies isotropic compressive stress on the cells. While other mechanical factors, such as shear stress and circumferential stretch, have been extensively studied, little is known about the role of hydrostatic pressure in the regulation of endothelial cell behavior. Here we show that hydrostatic pressure triggers partial and transient endothelial-to-mesenchymal transition in endothelial monolayers of different vascular beds. Values mimicking microvascular pressure environments promote proliferative and migratory behavior and impair barrier properties that are characteristic of a mesenchymal transition, resulting in increased sprouting angiogenesis in 3D organotypic model systems ex vivo and in vitro. Mechanistically, this response is linked to differential cadherin expression at the adherens junctions, and to an increased YAP expression, nuclear localization, and transcriptional activity. Inhibition of YAP transcriptional activity prevents pressure-induced sprouting angiogenesis. Together, this work establishes hydrostatic pressure as a key modulator of endothelial homeostasis and as a crucial component of the endothelial mechanical niche.
Collapse
Affiliation(s)
| | - Dominic Rütsche
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland
| | - Asra Abukar
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland
| | - Paul Hiebert
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
- Centre for Biomedicine, Hull York Medical School, The University of Hull, Hull, HU6 7RX, UK
| | - Dominik Zanetti
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
| | - Nikola Cesarovic
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zürich, Switzerland
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zürich, Switzerland
| | - Sabine Werner
- Department of Biology, ETH Zürich, Institute of Molecular Health Sciences, 8093, Zürich, Switzerland
| | - Edoardo Mazza
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland.
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland.
| | - Costanza Giampietro
- ETH Zürich, DMAVT, Experimental Continuum Mechanics, Zürich, 8092, Switzerland.
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Experimental Continuum Mechanics, Dübendorf, 8600, Switzerland.
| |
Collapse
|
21
|
Lechuga S, Marino-Melendez A, Davis A, Zalavadia A, Khan A, Longworth MS, Ivanov AI. Coactosin-like protein 1 regulates integrity and repair of model intestinal epithelial barriers via actin binding dependent and independent mechanisms. Front Cell Dev Biol 2024; 12:1405454. [PMID: 39040043 PMCID: PMC11260685 DOI: 10.3389/fcell.2024.1405454] [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: 03/22/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
The actin cytoskeleton regulates the integrity and repair of epithelial barriers by mediating the assembly of tight junctions (TJs), and adherens junctions (AJs), and driving epithelial wound healing. Actin filaments undergo a constant turnover guided by numerous actin-binding proteins, however, the roles of actin filament dynamics in regulating intestinal epithelial barrier integrity and repair remain poorly understood. Coactosin-like protein 1 (COTL1) is a member of the ADF/cofilin homology domain protein superfamily that binds and stabilizes actin filaments. COTL1 is essential for neuronal and cancer cell migration, however, its functions in epithelia remain unknown. The goal of this study is to investigate the roles of COTL1 in regulating the structure, permeability, and repair of the epithelial barrier in human intestinal epithelial cells (IEC). COTL1 was found to be enriched at apical junctions in polarized IEC monolayers in vitro. The knockdown of COTL1 in IEC significantly increased paracellular permeability, impaired the steady state TJ and AJ integrity, and attenuated junctional reassembly in a calcium-switch model. Consistently, downregulation of COTL1 expression in Drosophila melanogaster increased gut permeability. Loss of COTL1 attenuated collective IEC migration and decreased cell-matrix attachment. The observed junctional abnormalities in COTL1-depleted IEC were accompanied by the impaired assembly of the cortical actomyosin cytoskeleton. Overexpression of either wild-type COTL1 or its actin-binding deficient mutant tightened the paracellular barrier and activated junction-associated myosin II. Furthermore, the actin-uncoupled COTL1 mutant inhibited epithelial migration and matrix attachment. These findings highlight COTL1 as a novel regulator of the intestinal epithelial barrier integrity and repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
22
|
Müller L, Keil R, Glaß M, Hatzfeld M. Plakophilin 4 controls the spatio-temporal activity of RhoA at adherens junctions to promote cortical actin ring formation and tissue tension. Cell Mol Life Sci 2024; 81:291. [PMID: 38970683 PMCID: PMC11335210 DOI: 10.1007/s00018-024-05329-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/17/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024]
Abstract
Plakophilin 4 (PKP4) is a component of cell-cell junctions that regulates intercellular adhesion and Rho-signaling during cytokinesis with an unknown function during epidermal differentiation. Here we show that keratinocytes lacking PKP4 fail to develop a cortical actin ring, preventing adherens junction maturation and generation of tissue tension. Instead, PKP4-depleted cells display increased stress fibers. PKP4-dependent RhoA localization at AJs was required to activate a RhoA-ROCK2-MLCK-MLC2 axis and organize actin into a cortical ring. AJ-associated PKP4 provided a scaffold for the Rho activator ARHGEF2 and the RhoA effectors MLCK and MLC2, facilitating the spatio-temporal activation of RhoA signaling at cell junctions to allow cortical ring formation and actomyosin contraction. In contrast, association of PKP4 with the Rho suppressor ARHGAP23 reduced ARHGAP23 binding to RhoA which prevented RhoA activation in the cytoplasm and stress fiber formation. These data identify PKP4 as an AJ component that transduces mechanical signals into cytoskeletal organization.
Collapse
Affiliation(s)
- Lisa Müller
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany.
| | - René Keil
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany
| | - Mechthild Hatzfeld
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany
| |
Collapse
|
23
|
Suong DNA, Imamura K, Kato Y, Inoue H. Design of neural organoids engineered by mechanical forces. IBRO Neurosci Rep 2024; 16:190-195. [PMID: 38328799 PMCID: PMC10847990 DOI: 10.1016/j.ibneur.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
Neural organoids consist of three-dimensional tissue derived from pluripotent stem cells that could recapitulate key features of the human brain. During the past decade, organoid technology has evolved in the field of human brain science by increasing the quality and applicability of its products. Among them, a novel approach involving the design of neural organoids engineered by mechanical forces has emerged. This review describes previous approaches for the generation of neural organoids, the engineering of neural organoids by mechanical forces, and future challenges for the application of mechanical forces in the design of neural organoids.
Collapse
Affiliation(s)
- Dang Ngoc Anh Suong
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Keiko Imamura
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical‑Risk Avoidance Based On iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE MultiMix Corporation, Saitama, Japan
| | - Haruhisa Inoue
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical‑Risk Avoidance Based On iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| |
Collapse
|
24
|
Xie L, Wu G, Liu X, Duan X, Zhou K, Li H, Ning W. The TRIP6/LATS1 complex constitutes the tension sensor of α-catenin/vinculin at both bicellular and tricellular junctions. Eur J Cell Biol 2024; 103:151426. [PMID: 38805800 DOI: 10.1016/j.ejcb.2024.151426] [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: 11/29/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
Cell-cell mechanotransduction regulates tissue development and homeostasis. α-catenin, the core component of adherens junctions, functions as a tension sensor and transducer by recruiting vinculin and transducing signals that influence cell behaviors. α-catenin/vinculin complex-mediated mechanotransduction regulates multiple pathways, such as Hippo pathway. However, their associations with the α-catenin-based tension sensors at cell junctions are still not fully addressed. Here, we uncovered the TRIP6/LATS1 complex co-localizes with α-catenin/vinculin at both bicellular junctions (BCJs) and tricellular junctions (TCJs). The localization of TRIP6/LATS1 complex to both TCJs and BCJs requires ROCK1 and α-catenin. Treatment by cytochalasin B, Y-27632 and blebbistatin all impaired the BCJ and TCJ junctional localization of TRIP6/LATS1, indicating that the junctional localization of TRIP6/LATS1 is mechanosensitive. The α-catenin/vinculin/TRIP6/LATS1 complex strongly localized to TCJs and exhibited a discontinuous button-like pattern on BCJs. Additionally, we developed and validated an α-catenin/vinculin BiFC-based mechanosensor that co-localizes with TRIP6/LATS1 at BCJs and TCJs. The mechanosensor exhibited a discontinuous distribution and motile signals at BCJs. Overall, our study revealed that TRIP6 and LATS1 are novel compositions of the tension sensor, together with the core complex of α-catenin/vinculin, at both the BCJs and TCJs.
Collapse
Affiliation(s)
- Lin Xie
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Gangyun Wu
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Xiayu Liu
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Xiufen Duan
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Kaiyao Zhou
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Hua Li
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China.
| | - Wenxiu Ning
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China.
| |
Collapse
|
25
|
Li M, Xing X, Yuan J, Zeng Z. Research progress on the regulatory role of cell membrane surface tension in cell behavior. Heliyon 2024; 10:e29923. [PMID: 38720730 PMCID: PMC11076917 DOI: 10.1016/j.heliyon.2024.e29923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
Cell membrane surface tension has emerged as a pivotal biophysical factor governing cell behavior and fate. This review systematically delineates recent advances in techniques for cell membrane surface tension quantification, mechanosensing mechanisms, and regulatory roles of cell membrane surface tension in modulating major cellular processes. Micropipette aspiration, tether pulling, and newly developed fluorescent probes enable the measurement of cell membrane surface tension with spatiotemporal precision. Cells perceive cell membrane surface tension via conduits including mechanosensitive ion channels, curvature-sensing proteins (e.g. BAR domain proteins), and cortex-membrane attachment proteins (e.g. ERM proteins). Through membrane receptors like integrins, cells convert mechanical cues into biochemical signals. This conversion triggers cytoskeletal remodeling and extracellular matrix interactions in response to environmental changes. Elevated cell membrane surface tension suppresses cell spreading, migration, and endocytosis while facilitating exocytosis. Moreover, reduced cell membrane surface tension promotes embryonic stem cell differentiation and cancer cell invasion, underscoring cell membrane surface tension as a regulator of cell plasticity. Outstanding questions remain regarding cell membrane surface tension regulatory mechanisms and roles in tissue development/disease in vivo. Emerging tools to manipulate cell membrane surface tension with high spatiotemporal control in combination with omics approaches will facilitate the elucidation of cell membrane surface tension-mediated effects on signaling networks across various cell types/states. This will accelerate the development of cell membrane surface tension-based biomarkers and therapeutics for regenerative medicine and cancer. Overall, this review provides critical insights into cell membrane surface tension as a potent orchestrator of cell function, with broader impacts across mechanobiology.
Collapse
Affiliation(s)
- Manqing Li
- School of Public Health, Sun Yat-sen University, Guangzhou, 5180080, China
| | - Xiumei Xing
- School of Public Health, Sun Yat-sen University, Guangzhou, 5180080, China
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, 518054, China
| | - Zhuoying Zeng
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, 518035, China
- Chemical Analysis & Physical Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| |
Collapse
|
26
|
Nguyen TP, Otani T, Tsutsumi M, Kinoshita N, Fujiwara S, Nemoto T, Fujimori T, Furuse M. Tight junction membrane proteins regulate the mechanical resistance of the apical junctional complex. J Cell Biol 2024; 223:e202307104. [PMID: 38517380 PMCID: PMC10959758 DOI: 10.1083/jcb.202307104] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/29/2024] [Accepted: 02/16/2024] [Indexed: 03/23/2024] Open
Abstract
Epithelia must be able to resist mechanical force to preserve tissue integrity. While intercellular junctions are known to be important for the mechanical resistance of epithelia, the roles of tight junctions (TJs) remain to be established. We previously demonstrated that epithelial cells devoid of the TJ membrane proteins claudins and JAM-A completely lack TJs and exhibit focal breakages of their apical junctions. Here, we demonstrate that apical junctions fracture when claudin/JAM-A-deficient cells undergo spontaneous cell stretching. The junction fracture was accompanied by actin disorganization, and actin polymerization was required for apical junction integrity in the claudin/JAM-A-deficient cells. Further deletion of CAR resulted in the disruption of ZO-1 molecule ordering at cell junctions, accompanied by severe defects in apical junction integrity. These results demonstrate that TJ membrane proteins regulate the mechanical resistance of the apical junctional complex in epithelial cells.
Collapse
Affiliation(s)
- Thanh Phuong Nguyen
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Kawaguchi, Japan
| | - Motosuke Tsutsumi
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Noriyuki Kinoshita
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Sachiko Fujiwara
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tomomi Nemoto
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
27
|
López-Posadas R, Bagley DC, Pardo-Pastor C, Ortiz-Zapater E. The epithelium takes the stage in asthma and inflammatory bowel diseases. Front Cell Dev Biol 2024; 12:1258859. [PMID: 38529406 PMCID: PMC10961468 DOI: 10.3389/fcell.2024.1258859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
The epithelium is a dynamic barrier and the damage to this epithelial layer governs a variety of complex mechanisms involving not only epithelial cells but all resident tissue constituents, including immune and stroma cells. Traditionally, diseases characterized by a damaged epithelium have been considered "immunological diseases," and research efforts aimed at preventing and treating these diseases have primarily focused on immuno-centric therapeutic strategies, that often fail to halt or reverse the natural progression of the disease. In this review, we intend to focus on specific mechanisms driven by the epithelium that ensure barrier function. We will bring asthma and Inflammatory Bowel Diseases into the spotlight, as we believe that these two diseases serve as pertinent examples of epithelium derived pathologies. Finally, we will argue how targeting the epithelium is emerging as a novel therapeutic strategy that holds promise for addressing these chronic diseases.
Collapse
Affiliation(s)
- Rocío López-Posadas
- Department of Medicine 1, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universtiy Eralngen-Nürnberg, Erlangen, Germany
| | - Dustin C. Bagley
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Carlos Pardo-Pastor
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Universitat de Valencia, Valencia, Spain
- Instituto Investigación Hospital Clínico-INCLIVA, Valencia, Spain
| |
Collapse
|
28
|
Brenner LM, Meyer F, Yang H, Köhler AR, Bashtrykov P, Guo M, Jeltsch A, Lungu C, Olayioye MA. Repeat DNA methylation is modulated by adherens junction signaling. Commun Biol 2024; 7:286. [PMID: 38454140 PMCID: PMC10920906 DOI: 10.1038/s42003-024-05990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
Through its involvement in gene transcription and heterochromatin formation, DNA methylation regulates how cells interact with their environment. Nevertheless, the extracellular signaling cues that modulate the distribution of this central chromatin modification are largely unclear. DNA methylation is highly abundant at repetitive elements, but its investigation in live cells has been complicated by methodological challenges. Utilizing a CRISPR/dCas9 biosensor that reads DNA methylation of human α-satellite repeats in live cells, we here uncover a signaling pathway linking the chromatin and transcriptional state of repetitive elements to epithelial adherens junction integrity. Specifically, we find that in confluent breast epithelial cell monolayers, α-satellite repeat methylation is reduced by comparison to low density cultures. This is coupled with increased transcriptional activity at repeats. Through comprehensive perturbation experiments, we identify the junctional protein E-cadherin, which links to the actin cytoskeleton, as a central molecular player for signal relay into the nucleus. Furthermore, we find that this pathway is impaired in cancer cells that lack E-cadherin and are not contact-inhibited. This suggests that the molecular connection between cell density and repetitive element methylation could play a role in the maintenance of epithelial tissue homeostasis.
Collapse
Affiliation(s)
- Lisa-Marie Brenner
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Florian Meyer
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Haiqian Yang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Anja R Köhler
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Pavel Bashtrykov
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Albert Jeltsch
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Cristiana Lungu
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Nobelstraße 15, 70569, Stuttgart, Germany.
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Nobelstraße 15, 70569, Stuttgart, Germany.
| |
Collapse
|
29
|
Thomasy SM, Leonard BC, Greiner MA, Skeie JM, Raghunathan VK. Squishy matters - Corneal mechanobiology in health and disease. Prog Retin Eye Res 2024; 99:101234. [PMID: 38176611 PMCID: PMC11193890 DOI: 10.1016/j.preteyeres.2023.101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
The cornea, as a dynamic and responsive tissue, constantly interacts with mechanical forces in order to maintain its structural integrity, barrier function, transparency and refractive power. Cells within the cornea sense and respond to various mechanical forces that fundamentally regulate their morphology and fate in development, homeostasis and pathophysiology. Corneal cells also dynamically regulate their extracellular matrix (ECM) with ensuing cell-ECM crosstalk as the matrix serves as a dynamic signaling reservoir providing biophysical and biochemical cues to corneal cells. Here we provide an overview of mechanotransduction signaling pathways then delve into the recent advances in corneal mechanobiology, focusing on the interplay between mechanical forces and responses of the corneal epithelial, stromal, and endothelial cells. We also identify species-specific differences in corneal biomechanics and mechanotransduction to facilitate identification of optimal animal models to study corneal wound healing, disease, and novel therapeutic interventions. Finally, we identify key knowledge gaps and therapeutic opportunities in corneal mechanobiology that are pressing for the research community to address especially pertinent within the domains of limbal stem cell deficiency, keratoconus and Fuchs' endothelial corneal dystrophy. By furthering our understanding corneal mechanobiology, we can contextualize discoveries regarding corneal diseases as well as innovative treatments for them.
Collapse
Affiliation(s)
- Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States; California National Primate Research Center, Davis, CA, United States.
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States
| | - Mark A Greiner
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | - Jessica M Skeie
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | | |
Collapse
|
30
|
Shi Y, Li D, Yi B, Tang H, Xu T, Zhang Y. Physiological cyclic stretching potentiates the cell-cell junctions in vascular endothelial layer formed on aligned fiber substrate. BIOMATERIALS ADVANCES 2024; 157:213751. [PMID: 38219418 DOI: 10.1016/j.bioadv.2023.213751] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024]
Abstract
In vascular tissue engineering, formation of stable endothelial cell-cell and cell-substrate adhesions is essential for maintaining long-term patency of the tissue-engineered vascular grafts (TEVGs). In this study, sheet-like aligned fibrous substrates of poly(l-lactide-co-caprolactone) (PLCL) were prepared by electrospinning to provide basement membrane-resembling structural support to endothelial cells (ECs). Cyclic stretching at physiological and pathological levels was then applied to human umbilical vein endothelial cells (HUVECs) cultured on chosen fibrous substrate using a force-loading device, from which effects of the cyclic stretching on cell-cell and cell-substrate adhesions were examined. It was found that applying uniaxial 1 Hz cyclic stretch at physiological levels (5 % and 10 % elongation) strengthened the cell-cell junctions, thus leading to improved structural integrity, functional expression and resistance to thrombin-induced damaging impacts in the formed endothelial layer. The cell-cell junctions were disrupted at pathological level (15 % elongation) cyclic stretching, which however facilitated the formation of focal adhesions (FAs) at cell-substrate interface. Mechanistically, the effects of cyclic stretching on endothelial cell-cell and cell-substrate adhesions were identified to be correlated with the RhoA/ROCK signaling pathway. Results from this study highlight the relevance between applying dynamic mechanical stimulation and maintaining the structural integrity of the formed endothelial layer, and implicate a necessity to implement appropriate dynamic mechanical training (i.e., preconditioning) to obtain tissue-engineered blood vessels with long-term patency post-implantation.
Collapse
Affiliation(s)
- Yu Shi
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Donghong Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Bingcheng Yi
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Han Tang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Tingting Xu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Yanzhong Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China; Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, China.
| |
Collapse
|
31
|
Gao Q, Tian W, Yang H, Hu H, Zheng J, Yao X, Hu B, Liu H. Shen-Ling-Bai-Zhu-San alleviates the imbalance of intestinal homeostasis in dextran sodium sulfate-induced colitis mice by regulating gut microbiota and inhibiting the NLRP3 inflammasome activation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117136. [PMID: 37704122 DOI: 10.1016/j.jep.2023.117136] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shen-Ling-Bai-Zhu-San (SLBZS) is a classic formula for strengthening the spleen and dispelling dampness, which has shown excellent efficacy in inflammatory bowel disease (IBD) in traditional Chinese medicine clinical studies. However, its exact pharmacological mechanism needs to be further elucidated. AIM OF THE STUDY This study aims to investigate the restorative effect and mechanism of SLBZS on disturbed intestinal homeostasis in DSS-induced colitis mice. MATERIALS AND METHODS A colitis model was induced by 3% dextran sulfate sodium (DSS) for seven days, and SLBZS was administered by gavage. The influence of SLBZS on DSS-induced clinical symptoms and disease activity index (DAI) was monitored and analyzed. Alcian blue and fluorescein isothiocyanate-conjugated wheat germ agglutinin (FITC-WGA) staining were used to assess intestinal mucus changes. The expression of intestinal barrier function indexes and immune-associated indexes were determined by H&E staining, real-time quantitative PCR (RT-qPCR), and Western blot. And gut microbiota changes were detected by 16S rDNA sequencing technology. The antibiotic experiment was used to explore the role of gut microbiota in SLBZS treatment. RESULTS The results showed that SLBZS significantly improved the physiological indexes including body weight, DAI score, and colon length of colitis mice. We focused on the effects of SLBZS on intestinal homeostasis in colitis mice. First, SLBZS could enhance the secretion of intestinal mucin and the expression levels of tight junctions and adhesive junctions. Second, SLBZS inhibited the expression level of inflammatory factors and reduced the protein expression level of NLRP3 inflammasome. Third, 16S rDNA sequencing analysis revealed that SLBZS repaired the dysfunctional gut microbiota of colitis mice, such as enhancing the abundance of short-chain fatty acid-producing bacteria including Faecalibaculum, Colidextribacter, and Coprococcus. Further, by gut microbiota-depleted mice, we found that SLBZS could not exert an anti-colitis effect when gut microbiota was absent. CONCLUSIONS SLBZS restored intestinal environmental homeostasis by enhancing intestinal barrier function, inhibiting NLRP3 inflammasome, and restoring disturbed gut microbiota. And SLBZS could not ameliorate colitis mice with depleted gut microbiota. Our finding provided a theoretical basis for the clinical application of SLBZS in IBD.
Collapse
Affiliation(s)
- Qianru Gao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| | - Weiyi Tian
- College of Basic Medical Sciences, Guizhou University of Traditional Chinese Medicine, Dongqing Road 4, Guiyang, 550025, PR China.
| | - Huabing Yang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| | - Haiming Hu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| | - Junping Zheng
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| | - Xiaowei Yao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| | - Baifei Hu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| | - Hongtao Liu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| |
Collapse
|
32
|
Xu Z, Xu X, Yang B, Mi Y, Wang J. 3D sheep rumen epithelial structures driven from single cells in vitro. Vet Res 2023; 54:104. [PMID: 37946298 PMCID: PMC10636852 DOI: 10.1186/s13567-023-01234-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/22/2023] [Indexed: 11/12/2023] Open
Abstract
Ruminants play a vital economic role as livestock, providing high-quality protein for humans. At present, 3D-cultured ruminant abomasum and intestinal organoids have been successfully established to study host and pathogen interaction. The rumen is a unique digestive organ of ruminants that occupies 70% of the volume of the digestive tract and its microbiota can decompose lignocellulose to support animal growth. Here we report a method for culturing rumen epithelial organoids. We found that single rumen epithelial cells form self-organized 3D structures representative of typical stratified squamous epithelium, which is similar to rumen epithelium. EGF, Noggin, Wnt3a, IGF-1, and FGF-10 significantly enhanced the seeding efficiency of organoids. Moreover, the inclusion of CHIR-99021, A83-01, SB202190, and Y-27632 is crucial for organoid formation and maintenance. Importantly, we demonstrate that rumen epithelial cells retain their ability to form organoids after passage, cryopreservation, and resuscitation. The rumen epithelial organoids express rumen cell type-specific genes, uptake fatty acids, and generate 2D cultures. In summary, our data demonstrate that it is feasible to establish organoids from single rumen epithelial cells, which is a novel in vitro system that may reduce the use of experimental animals.
Collapse
Affiliation(s)
- Zebang Xu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Xinxin Xu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Bin Yang
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, 310023, Zhejiang, China
| | - Yuling Mi
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiakun Wang
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- MoE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China.
| |
Collapse
|
33
|
Haferkamp U, Hartmann C, Abid CL, Brachner A, Höchner A, Gerhartl A, Harwardt B, Leckzik S, Leu J, Metzger M, Nastainczyk-Wulf M, Neuhaus W, Oerter S, Pless O, Rujescu D, Jung M, Appelt-Menzel A. Human isogenic cells of the neurovascular unit exert transcriptomic cell type-specific effects on a blood-brain barrier in vitro model of late-onset Alzheimer disease. Fluids Barriers CNS 2023; 20:78. [PMID: 37907966 PMCID: PMC10617216 DOI: 10.1186/s12987-023-00471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/01/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND The function of the blood-brain barrier (BBB) is impaired in late-onset Alzheimer disease (LOAD), but the associated molecular mechanisms, particularly with respect to the high-risk APOE4/4 genotype, are not well understood. For this purpose, we developed a multicellular isogenic model of the neurovascular unit (NVU) based on human induced pluripotent stem cells. METHODS The human NVU was modeled in vitro using isogenic co-cultures of astrocytes, brain capillary endothelial-like cells (BCECs), microglia-like cells, neural stem cells (NSCs), and pericytes. Physiological and pathophysiological properties were investigated as well as the influence of each single cell type on the characteristics and function of BCECs. The barriers established by BCECs were analyzed for specific gene transcription using high-throughput quantitative PCR. RESULTS Co-cultures were found to tighten the barrier of BCECs and alter its transcriptomic profile under both healthy and disease conditions. In vitro differentiation of brain cell types that constitute the NVU was not affected by the LOAD background. The supportive effect of NSCs on the barrier established by BCECs was diminished under LOAD conditions. Transcriptomes of LOAD BCECs were modulated by different brain cell types. NSCs were found to have the strongest effect on BCEC gene regulation and maintenance of the BBB. Co-cultures showed cell type-specific functional contributions to BBB integrity under healthy and LOAD conditions. CONCLUSIONS Cell type-dependent transcriptional effects on LOAD BCECs were identified. Our study suggests that different brain cell types of the NVU have unique roles in maintaining barrier integrity that vary under healthy and LOAD conditions. .
Collapse
Affiliation(s)
- Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525, Hamburg, Germany
| | - Carla Hartmann
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Chaudhry Luqman Abid
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Andreas Brachner
- Center Health and Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Vienna, 1210, Austria
| | - Alevtina Höchner
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany
| | - Anna Gerhartl
- Center Health and Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Vienna, 1210, Austria
| | - Bernadette Harwardt
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Selin Leckzik
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Jennifer Leu
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525, Hamburg, Germany
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
| | | | - Winfried Neuhaus
- Center Health and Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Vienna, 1210, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, 3500, Austria
| | - Sabrina Oerter
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525, Hamburg, Germany
| | - Dan Rujescu
- Department of Psychiatry and Psychotherapy, Division of General Psychiatry, Medical University of Vienna, Vienna, 1090, Austria
| | - Matthias Jung
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany.
| | - Antje Appelt-Menzel
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany.
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany.
| |
Collapse
|
34
|
Gou J, Zhang T, Othmer HG. The Interaction of Mechanics and the Hippo Pathway in Drosophila melanogaster. Cancers (Basel) 2023; 15:4840. [PMID: 37835534 PMCID: PMC10571775 DOI: 10.3390/cancers15194840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/10/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
Drosophila melanogaster has emerged as an ideal system for studying the networks that control tissue development and homeostasis and, given the similarity of the pathways involved, controlled and uncontrolled growth in mammalian systems. The signaling pathways used in patterning the Drosophila wing disc are well known and result in the emergence of interaction of these pathways with the Hippo signaling pathway, which plays a central role in controlling cell proliferation and apoptosis. Mechanical effects are another major factor in the control of growth, but far less is known about how they exert their control. Herein, we develop a mathematical model that integrates the mechanical interactions between cells, which occur via adherens and tight junctions, with the intracellular actin network and the Hippo pathway so as to better understand cell-autonomous and non-autonomous control of growth in response to mechanical forces.
Collapse
Affiliation(s)
- Jia Gou
- Department of Mathematics, University of California, Riverside, CA 92507, USA;
| | - Tianhao Zhang
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Hans G. Othmer
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
35
|
Jensen OE, Revell CK. Couple stresses and discrete potentials in the vertex model of cellular monolayers. Biomech Model Mechanobiol 2023; 22:1465-1486. [PMID: 36201070 PMCID: PMC10511640 DOI: 10.1007/s10237-022-01620-2] [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: 05/20/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022]
Abstract
The vertex model is widely used to simulate the mechanical properties of confluent epithelia and other multicellular tissues. This inherently discrete framework allows a Cauchy stress to be attributed to each cell, and its symmetric component has been widely reported, at least for planar monolayers. Here, we consider the stress attributed to the neighbourhood of each tricellular junction, evaluating in particular its leading-order antisymmetric component and the associated couple stresses, which characterise the degree to which individual cells experience (and resist) in-plane bending deformations. We develop discrete potential theory for localised monolayers having disordered internal structure and use this to derive the analogues of Airy and Mindlin stress functions. These scalar potentials typically have broad-banded spectra, highlighting the contributions of small-scale defects and boundary layers to global stress patterns. An affine approximation attributes couple stresses to pressure differences between cells sharing a trijunction, but simulations indicate an additional role for non-affine deformations.
Collapse
Affiliation(s)
- Oliver E. Jensen
- Department of Mathematics, University of Manchester, Oxford Road, Manchester, M13 9PL UK
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester, M13 9PL UK
| | - Christopher K. Revell
- Department of Mathematics, University of Manchester, Oxford Road, Manchester, M13 9PL UK
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester, M13 9PL UK
| |
Collapse
|
36
|
Labarrade F, Perrin A, Ferreira Y, Botto JM, Imbert I. Modulation of Piezo1 influences human skin architecture and oxytocin expression. Int J Cosmet Sci 2023; 45:604-611. [PMID: 37170671 DOI: 10.1111/ics.12864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/13/2023]
Abstract
OBJECTIVE Throughout our existence, the skin senses and analyses the mechanical forces imposed by the environment. In response to these environmental forces, skin can deform itself and achieve a biological response. The subsequent cutaneous plasticity emerges from mechanical properties arising from the collective action of the skin cells, particularly keratinocytes, that govern the tensile strength via cell-to-cell adhesions and via cell-matrix adhesion structures. In addition to serving as force-bearing entities, keratinocytes respond to forces by activating signalling pathways to control their own fate and function. To detect and adapt to mechanical signals, keratinocytes possess a panel of sensory receptors and junctional intercellular structures. Mechanically activated ion channel Piezo1 has been described as a force sensor and as being involved in pleasant touch perception. In this study, relationships between Piezo1 modulation and oxytocin synthesis were investigated. METHODS The expression of Piezo1 in the skin was studied and compared with the expression of TRPV1. Dooku1 antagonist and Jedi1 agonist were used to modulate Piezo1. The level of E-cadherin and oxytocin was monitored in ex vivo skin biopsies by immunodetection. RESULTS Taken together, our results illustrate the major role of mechanosensitive ion channel Piezo1 in skin barrier integrity, and in peripheral oxytocin synthesis in the skin. CONCLUSION In conclusion, this study highlights the relationships between pleasant touch, soft touch and local oxytocin synthesis.
Collapse
Affiliation(s)
| | - Armelle Perrin
- Ashland Global Skin Research Centre, Sophia Antipolis, France
| | - Yolène Ferreira
- Ashland Global Skin Research Centre, Sophia Antipolis, France
| | | | - Isabelle Imbert
- Ashland Global Skin Research Centre, Sophia Antipolis, France
| |
Collapse
|
37
|
Lei Y, Sun Y, Wu W, Liu H, Wang X, Shu Y, Fang S. Influenza H7N9 virus disrupts the monolayer human brain microvascular endothelial cells barrier in vitro. Virol J 2023; 20:219. [PMID: 37773164 PMCID: PMC10541704 DOI: 10.1186/s12985-023-02163-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/16/2023] [Indexed: 10/01/2023] Open
Abstract
Influenza H7N9 virus causes human infections with about 40% case fatality rate. The severe cases usually present with pneumonia; however, some present with central nervous system complications. Pneumonia syndrome is attributed to the cytokine storm after infection with H7N9, but the pathogenic mechanism of central nervous system complications has not been clarified. This study used immortalized human brain microvascular endothelial cells hCMEC/D3 to simulate the blood-brain barrier. It demonstrated that H7N9 virus could infect brain microvascular endothelial cells and compromise the blood-brain barrier integrity and permeability by down-regulating the expression of cell junction-related proteins, including claudin-5, occludin, and vascular endothelial (VE)-cadherin. These results suggested that H7N9 could infect the blood-brain barrier in vitro and affect its functions, which could be a potential mechanism for the pathogenesis of H7N9 viral encephalopathy.
Collapse
Affiliation(s)
- Yuxuan Lei
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Ying Sun
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Weihua Wu
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Hui Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Xin Wang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Shisong Fang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
| |
Collapse
|
38
|
Han H, Ke L, Xu W, Wang H, Zhou J, Rao P. Incidental nanoparticles in black tea alleviate DSS-induced ulcerative colitis in BALB/c mice. Food Funct 2023; 14:8420-8430. [PMID: 37615587 DOI: 10.1039/d3fo00641g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
As the dominant herbal drink consumed worldwide, black tea exhibits various health promoting benefits including amelioration of inflammatory bowel diseases. Despite extensive studies on the tea's components, little is known about the bioactivities of nanoparticles (NPs) which were incidentally assembled in the tea infusion and represent the major components. This study investigated the alleviative effects of black tea infusion, the isolated black tea NPs, and a mixture of caffeine, epigallocatechin-3-gallate, gallic acid and epicatechin gallate on dextran sodium sulfate (DSS)-induced ulcerative colitis. The results showed that both the black tea infusion and the NPs significantly alleviated colitis, suppressed the mRNA levels of pro-inflammatory cytokines TNF-α, IL-6, and IL-1β, and suppressed the DSS-induced loss of cell-cell junction proteins (e.g., E-cadherin, ZO-1, and claudin-1) and increase of p-STAT3. The mixture of four tea components, which is the analogue of bioactive payloads carried by the NPs, was much less effective than the tea infusion and NPs. It shows that the NPs elevate the efficiency of polyphenols and caffeine in black tea in restoring the intercellular connection in the intestine, inhibiting mucosal inflammation, and alleviating ulcerative colitis. This work may inspire the development of tea-based therapeutics for treating inflammatory bowel diseases and have wide influences on value-added processing, quality evaluation, functionalization, and innovation of tea and other plant-based beverages.
Collapse
Affiliation(s)
- Huan Han
- School of Chemical Engineering and Technology, Tianjin University, China
- Zhe Jiang Institute of Tianjin University, Shaoxing, China
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Lijing Ke
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310012, China
- School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT, UK.
| | - Wei Xu
- Zhe Jiang Institute of Tianjin University, Shaoxing, China
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Huiqin Wang
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Jianwu Zhou
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Pingfan Rao
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310012, China
| |
Collapse
|
39
|
Ke W, Liao Z, Liang H, Tong B, Song Y, Li G, Ma L, Wang K, Feng X, Li S, Hua W, Wang B, Yang C. Stiff Substrate Induces Nucleus Pulposus Cell Ferroptosis via YAP and N-Cadherin Mediated Mechanotransduction. Adv Healthc Mater 2023; 12:e2300458. [PMID: 37022980 DOI: 10.1002/adhm.202300458] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/26/2023] [Indexed: 04/07/2023]
Abstract
Increased tissue stiffness is associated with various pathological processes, such as fibrosis, inflammation, and aging. The matrix stiffness of the nucleus pulposus (NP) tissues increases gradually during intervertebral disc degeneration (IDD), while the mechanism through which NP cells sense and react to matrix stiffness remains unclear. In this study, the results indicate that ferroptosis is involved in stiff substrate-induced NP cell death. The expression of acyl-CoA synthetase long-chain family member 4 (ACSL4) increases in NP cells of the stiff group, which mediates lipid peroxidation and ferroptosis in NP cells. In addition, stiff substrate activates the hippo signaling cascade and induces the nuclear translocation of yes-associated protein (YAP). Interestingly, inhibition of YAP is efficient to reverse the increase of ACSL4 expression caused by matrix stiffness. Furthermore, stiff substrate suppresses the expression of N-cadherin in NP cells. N-cadherin overexpression can inhibit YAP nuclear translocation via the formation of the N-cadherin/β-catenin/YAP complex, and reverse matrix stiffness-induced ferroptosis in NP cells. Finally, the effects of YAP inhibition and N-cadherin overexpression on IDD progression are further illustrated in animal models. These findings reveal a new mechanism of mechanotransduction in NP cells, providing novel insights into the development of therapies for the treatment of IDD.
Collapse
Affiliation(s)
- Wencan Ke
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bide Tong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bingjin Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
40
|
Cui W, Subramani A, Fonseca P, Zhang Y, Tong L, Zhang Y, Egevad L, Lundqvist A, Holmgren L. Deciphering the Role of p60AmotL2 in Epithelial Extrusion and Cell Detachment. Cells 2023; 12:2158. [PMID: 37681890 PMCID: PMC10486482 DOI: 10.3390/cells12172158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 09/09/2023] Open
Abstract
Preserving an accurate cell count is crucial for maintaining homeostasis. Apical extrusion, a process in which redundant cells are eliminated by neighboring cells, plays a key role in this regard. Recent studies have revealed that apical extrusion can also be triggered in cells transformed by oncogenes, suggesting it may be a mechanism through which tumor cells escape their microenvironment. In previous work, we demonstrated that p60AmotL2 modulates the E-cadherin function by inhibiting its connection to radial actin filaments. This isoform of AmotL2 is expressed in invasive breast and colon tumors and promotes invasion in vitro and in vivo. Transcriptionally regulated by c-Fos, p60AmotL2 is induced by local stress signals such as severe hypoxia. In this study, we investigated the normal role of p60AmotL2 in epithelial tissues. We found that this isoform is predominantly expressed in the gut, where cells experience rapid turnover. Through time-lapse imaging, we present evidence that cells expressing p60AmotL2 are extruded by their normal neighboring cells. Based on these findings, we hypothesize that tumor cells exploit this pathway to detach from normal epithelia and invade surrounding tissues.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lars Holmgren
- Department of Oncology-Pathology, Bioclinicum J6:20, Solnavägen 30, Karolinska Institutet, 171 64 Stockholm, Sweden (L.E.)
| |
Collapse
|
41
|
He J, Xie X, Xiao Z, Qian W, Zhang L, Hou X. Piezo1 in Digestive System Function and Dysfunction. Int J Mol Sci 2023; 24:12953. [PMID: 37629134 PMCID: PMC10454946 DOI: 10.3390/ijms241612953] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Piezo1, a non-selective cation channel directly activated by mechanical forces, is widely expressed in the digestive system and participates in biological functions physiologically and pathologically. In this review, we summarized the latest insights on Piezo1's cellular effect across the entire digestive system, and discussed the role of Piezo1 in various aspects including ingestion and digestion, material metabolism, enteric nervous system, intestinal barrier, and inflammatory response within digestive system. The goal of this comprehensive review is to provide a solid foundation for future research about Piezo1 in digestive system physiologically and pathologically.
Collapse
Affiliation(s)
| | | | | | | | - Lei Zhang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.H.); (X.X.); (Z.X.); (W.Q.)
| | - Xiaohua Hou
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.H.); (X.X.); (Z.X.); (W.Q.)
| |
Collapse
|
42
|
Mahlandt EK, Palacios Martínez S, Arts JJG, Tol S, van Buul JD, Goedhart J. Opto-RhoGEFs, an optimized optogenetic toolbox to reversibly control Rho GTPase activity on a global to subcellular scale, enabling precise control over vascular endothelial barrier strength. eLife 2023; 12:RP84364. [PMID: 37449837 PMCID: PMC10393062 DOI: 10.7554/elife.84364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The inner layer of blood vessels consists of endothelial cells, which form the physical barrier between blood and tissue. This vascular barrier is tightly regulated and is defined by cell-cell contacts through adherens and tight junctions. To investigate the signaling that regulates vascular barrier strength, we focused on Rho GTPases, regulators of the actin cytoskeleton and known to control junction integrity. To manipulate Rho GTPase signaling in a temporal and spatial manner we applied optogenetics. Guanine-nucleotide exchange factor (GEF) domains from ITSN1, TIAM1, and p63RhoGEF, activating Cdc42, Rac, and Rho, respectively, were integrated into the optogenetic recruitment tool improved light-induced dimer (iLID). This tool allows for Rho GTPase activation at the subcellular level in a reversible and non-invasive manner by recruiting a GEF to a specific area at the plasma membrane, The membrane tag of iLID was optimized and a HaloTag was applied to gain more flexibility for multiplex imaging. The resulting optogenetically recruitable RhoGEFs (Opto-RhoGEFs) were tested in an endothelial cell monolayer and demonstrated precise temporal control of vascular barrier strength by a cell-cell overlap-dependent, VE-cadherin-independent, mechanism. Furthermore, Opto-RhoGEFs enabled precise optogenetic control in endothelial cells over morphological features such as cell size, cell roundness, local extension, and cell contraction. In conclusion, we have optimized and applied the optogenetic iLID GEF recruitment tool, that is Opto-RhoGEFs, to study the role of Rho GTPases in the vascular barrier of the endothelium and found that membrane protrusions at the junction region can rapidly increase barrier integrity independent of VE-cadherin.
Collapse
Affiliation(s)
- Eike K Mahlandt
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, Netherlands
| | - Sebastián Palacios Martínez
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, Netherlands
| | - Janine J G Arts
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, Netherlands
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| | - Simon Tol
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| | - Jaap D van Buul
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, Netherlands
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| | - Joachim Goedhart
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
43
|
Liu Q, Sun Y, Guan L, Chen X, Zhou J, Liu P, Huo B. Detection of the effect of microvibrational stimulation on human discarded immature oocytes by single-cell transcriptome sequencing technology. J Assist Reprod Genet 2023; 40:1773-1781. [PMID: 37273164 PMCID: PMC10352214 DOI: 10.1007/s10815-023-02837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/23/2023] [Indexed: 06/06/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the changes in oocytes at the transcriptome level after applying continuous microvibrational mechanical stimulation to human immature oocytes during in vitro maturation. METHODS The discarded germinal-vesicle stage (GV) oocytes with no fertilization value after oocytes retrieval in assisted reproduction cycles were collected. Part of them was stimulated with vibration (n = 6) at 10 Hz for 24 h after obtaining informed consent; the other was cultured in static condition (n = 6). Single-cell transcriptome sequencing was used to detect the differences in oocyte transcriptome compared with the static culture group. RESULTS The applied 10-Hz continuous microvibrational stimulation altered the expression of 352 genes compared with the static culture. Gene Ontology (GO) analysis suggested that the altered genes were mainly enriched with 31 biological processes. The mechanical stimulation upregulated 155 of these genes and downregulated 197 genes. Among them, the genes related to mechanical signaling, such as protein localization to intercellular adhesion (DSP and DLG-5) and cytoskeleton (DSP, FGD6, DNAJC7, KRT16, KLHL1, HSPB1, MAP2K6), were detected. DLG-5, which was related to protein localization to intercellular adhesion, was selected for immunofluorescence experiments based on the transcriptome sequencing results. The protein expression of DLG-5 in the microvibration-stimulated oocytes was higher than that in the static culture oocytes. CONCLUSIONS Mechanical stimulation affects the transcriptome during oocyte maturation, causing the express changes in intercellular adhesion and cytoskeleton-related genes. We speculate that the mechanical signal may be transmitted to the cell through DLG-5 protein and cytoskeleton-related protein to regulate cellular activities.
Collapse
Affiliation(s)
- Qinli Liu
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| | - Yanxia Sun
- Reproductive Medical Center, Amcare Women's & Children's Hospital, Tianjin, China
| | - Lijun Guan
- Reproductive Medical Center, Amcare Women's & Children's Hospital, Tianjin, China
| | - Xinna Chen
- Reproductive Medical Center, Amcare Women's & Children's Hospital, Tianjin, China
| | - Jian Zhou
- Reproductive Medical Center, Amcare Women's & Children's Hospital, Tianjin, China
| | - Ping Liu
- Department of Obstetrics and Gynecology, Reproductive Medical Centre, Peking University Third Hospital, Beijing, China
| | - Bo Huo
- Sport Biomechanics Center, Sports Artificial Intelligence Institute, Capital University of Physical Education and Sports, Beijing, 100191, China.
| |
Collapse
|
44
|
Yu X, He L, Lin W, Zheng X, Zhang L, Yu B, Wang Y, Yang Z, Lin Y. Long-term menopause exacerbates vaginal wall support injury in ovariectomized rats by regulating amino acid synthesis and glycerophospholipid metabolism. Front Endocrinol (Lausanne) 2023; 14:1119599. [PMID: 37424873 PMCID: PMC10324610 DOI: 10.3389/fendo.2023.1119599] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Purpose Menopause is a risk factor for pelvic organ prolapse (POP) and is frequently associated with diminished vaginal wall support. To uncover relevant molecular mechanisms and provide potential therapeutic targets, we evaluated changes in the transcriptome and metabolome of the vaginal wall in ovariectomized rats to identify important molecular changes. Methods Sixteen adult female Sprague-Dawley rats were randomly assigned to either the control or menopause group. Seven months after the operation, hematoxylin and eosin (H&E) staining and Masson trichrome staining were used to observe changes in the rat vaginal wall structure. Differentially expressed genes (DEGs) and metabolites (DEMs) in the vaginal wall were detected by RNA-sequencing and LC-MS, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of DEGs and DEMs were performed. Results We verified that long-term menopause causes vaginal wall injury by H&E and Masson trichrome staining. From the multiomics analyses, 20,669 genes and 2193 metabolites were identified. Compared with the control group, 3255 DEGs were found in the vaginal wall of long-term menopausal rats. Bioinformatics analysis showed that the DEGs were mainly enriched in mechanistic pathways, including cell-cell junction, extracellular matrix, muscle tissue developments, the PI3K-Akt signaling pathway, the MAPK signaling pathway, tight junctions and the Wnt signaling pathway. Additionally, 313 DEMs were found, and they consisted mostly of amino acids and their metabolites. DEMs were also enriched in mechanistic pathways, such as glycine, serine and threonine metabolism, glycerophospholipid metabolism, gap junctions and ferroptosis. Coexpression analysis of DEGs and DEMs revealed that biosynthesis of amino acids (isocitric acid and PKM) and glycerophospholipid metabolism (1-(9Z-hexadecenoyl)-sn-glycero-3-phosphocholine and PGS1) are critical metabolic pathways, suggesting that POP induced by menopause may be associated with the regulation of these processes. Conclusion The findings showed that long-term menopause greatly exacerbated vaginal wall support injury by decreasing the biosynthesis of amino acids and interfering with glycerophospholipid metabolism, which may result in POP. This study not only clarified that long-term menopause exacerbates damage to the vaginal wall but also provided insight into the potential molecular mechanisms by which long-term menopause induces POP.
Collapse
Affiliation(s)
- Xia Yu
- Department of Clinical Laboratory, Chengdu Women’s and Children’s Central Hospital, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Li He
- Department of Obstetrics and Gynecology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Wenyi Lin
- Department of Medical Pathology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xuemei Zheng
- Department of Obstetrics and Gynecology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ling Zhang
- Department of Obstetrics and Gynecology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bo Yu
- Department of Medical Pathology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yanjun Wang
- Department of Obstetrics and Gynecology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yonghong Lin
- Department of Obstetrics and Gynecology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Subramani A, Cui W, Zhang Y, Friman T, Zhao Z, Huang W, Fonseca P, Lui WO, Narayanan V, Bobrowska J, Lekka M, Yan J, Conway DE, Holmgren L. Modulation of E-Cadherin Function through the AmotL2 Isoforms Promotes Ameboid Cell Invasion. Cells 2023; 12:1682. [PMID: 37443716 PMCID: PMC10340588 DOI: 10.3390/cells12131682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 07/15/2023] Open
Abstract
The spread of tumor cells and the formation of distant metastasis remain the main causes of mortality in cancer patients. However, the mechanisms governing the release of cells from micro-environmental constraints remain unclear. E-cadherin negatively controls the invasion of epithelial cells by maintaining cell-cell contacts. Furthermore, the inactivation of E-cadherin triggers invasion in vitro. However, the role of E-cadherin is complex, as metastasizing cells maintain E-cadherin expression, which appears to have a positive role in the survival of tumor cells. In this report, we present a novel mechanism delineating how E-cadherin function is modulated to promote invasion. We have previously shown that E-cadherin is associated with p100AmotL2, which is required for radial actin formation and the transmission of mechanical force. Here, we present evidence that p60AmotL2, which is expressed in invading tumor cells, binds to the p100AmotL2 isoform and uncouples the mechanical constraint of radial actin filaments. We show for the first time that the coupling of E-cadherin to the actin cytoskeleton via p100AmotL2 is directly connected to the nuclear membrane. The expression of p60AmotL2 inactivates this connection and alters the properties of the nuclear lamina, potentiating the invasion of cells into micropores of the extracellular matrix. In summary, we propose that the balance of the two AmotL2 isoforms is important in the modulation of E-cadherin function and that an imbalance of this axis promotes ameboid cell invasion.
Collapse
Affiliation(s)
- Aravindh Subramani
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Weiyingqi Cui
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Yuanyuan Zhang
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Tomas Friman
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Zhihai Zhao
- Department of Physics, Faculty of Science: 2 Science Drive 3, S7-01-10, Lower Kent Ridge Road, Singapore 117542, Singapore; (Z.Z.); (W.H.); (J.Y.)
- Mechanobiology Institute (MBI): T-Lab, #10-02, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Wenmao Huang
- Department of Physics, Faculty of Science: 2 Science Drive 3, S7-01-10, Lower Kent Ridge Road, Singapore 117542, Singapore; (Z.Z.); (W.H.); (J.Y.)
- Mechanobiology Institute (MBI): T-Lab, #10-02, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Pedro Fonseca
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Weng-Onn Lui
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 West Main Street, Richmond, VA 23284, USA; (V.N.); (D.E.C.)
| | - Justyna Bobrowska
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland; (J.B.); (M.L.)
| | - Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland; (J.B.); (M.L.)
| | - Jie Yan
- Department of Physics, Faculty of Science: 2 Science Drive 3, S7-01-10, Lower Kent Ridge Road, Singapore 117542, Singapore; (Z.Z.); (W.H.); (J.Y.)
- Mechanobiology Institute (MBI): T-Lab, #10-02, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Daniel E. Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 West Main Street, Richmond, VA 23284, USA; (V.N.); (D.E.C.)
| | - Lars Holmgren
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| |
Collapse
|
46
|
Shi B, Matsui T, Qian S, Weiss TM, Nicholl ID, Callaway DJE, Bu Z. An ensemble of cadherin-catenin-vinculin complex employs vinculin as the major F-actin binding mode. Biophys J 2023; 122:2456-2474. [PMID: 37147801 PMCID: PMC10323030 DOI: 10.1016/j.bpj.2023.04.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/14/2023] [Accepted: 04/27/2023] [Indexed: 05/07/2023] Open
Abstract
The cell-cell adhesion cadherin-catenin complexes recruit vinculin to the adherens junction (AJ) to modulate the mechanical couplings between neighboring cells. However, it is unclear how vinculin influences the AJ structure and function. Here, we identified two patches of salt bridges that lock vinculin in the head-tail autoinhibited conformation and reconstituted the full-length vinculin activation mimetics bound to the cadherin-catenin complex. The cadherin-catenin-vinculin complex contains multiple disordered linkers and is highly dynamic, which poses a challenge for structural studies. We determined the ensemble conformation of this complex using small-angle x-ray and selective deuteration/contrast variation small-angle neutron scattering. In the complex, both α-catenin and vinculin adopt an ensemble of flexible conformations, but vinculin has fully open conformations with the vinculin head and actin-binding tail domains well separated from each other. F-actin binding experiments show that the cadherin-catenin-vinculin complex binds and bundles F-actin. However, when the vinculin actin-binding domain is removed from the complex, only a minor fraction of the complex binds to F-actin. The results show that the dynamic cadherin-catenin-vinculin complex employs vinculin as the primary F-actin binding mode to strengthen AJ-cytoskeleton interactions.
Collapse
Affiliation(s)
- Bright Shi
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York; PhD Programs in Chemistry and Biochemistry, CUNY Graduate Center, New York
| | - Tsutomu Matsui
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | - Shuo Qian
- Second Target Station Project, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Thomas M Weiss
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | - Iain D Nicholl
- Department of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom
| | - David J E Callaway
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York.
| | - Zimei Bu
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York; PhD Programs in Chemistry and Biochemistry, CUNY Graduate Center, New York.
| |
Collapse
|
47
|
Capaldo CT. Claudin Barriers on the Brink: How Conflicting Tissue and Cellular Priorities Drive IBD Pathogenesis. Int J Mol Sci 2023; 24:8562. [PMID: 37239907 PMCID: PMC10218714 DOI: 10.3390/ijms24108562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are characterized by acute or chronic recurring inflammation of the intestinal mucosa, often with increasing severity over time. Life-long morbidities and diminishing quality of life for IBD patients compel a search for a better understanding of the molecular contributors to disease progression. One unifying feature of IBDs is the failure of the gut to form an effective barrier, a core role for intercellular complexes called tight junctions. In this review, the claudin family of tight junction proteins are discussed as they are a fundamental component of intestinal barriers. Importantly, claudin expression and/or protein localization is altered in IBD, leading to the supposition that intestinal barrier dysfunction exacerbates immune hyperactivity and disease. Claudins are a large family of transmembrane structural proteins that constrain the passage of ions, water, or substances between cells. However, growing evidence suggests non-canonical claudin functions during mucosal homeostasis and healing after injury. Therefore, whether claudins participate in adaptive or pathological IBD responses remains an open question. By reviewing current studies, the possibility is assessed that with claudins, a jack-of-all-trades is master of none. Potentially, a robust claudin barrier and wound restitution involve conflicting biophysical phenomena, exposing barrier vulnerabilities and a tissue-wide frailty during healing in IBD.
Collapse
Affiliation(s)
- Christopher T Capaldo
- College of Natural and Computer Sciences, Hawai'i Pacific University, Honolulu, HI 96813, USA
| |
Collapse
|
48
|
Troyanovsky SM. Adherens junction: the ensemble of specialized cadherin clusters. Trends Cell Biol 2023; 33:374-387. [PMID: 36127186 PMCID: PMC10020127 DOI: 10.1016/j.tcb.2022.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022]
Abstract
The cell-cell connections in adherens junctions (AJs) are mediated by transmembrane receptors, type I cadherins (referred to here as cadherins). These cadherin-based connections (or trans bonds) are weak. To upregulate their strength, cadherins exploit avidity, the increased affinity of binding between cadherin clusters compared with isolated monomers. Formation of such clusters is a unique molecular process that is driven by a synergy of direct and indirect cis interactions between cadherins located at the same cell. In addition to their role in adhesion, cadherin clusters provide structural scaffolds for cytosolic proteins, which implicate cadherin into different cellular activities and signaling pathways. The cluster lifetime, which depends on the actin cytoskeleton, and on the mechanical forces it generates, determines the strength of AJs and their plasticity. The key aspects of cadherin adhesion, therefore, cannot be understood at the level of isolated cadherin molecules, but should be discussed in the context of cadherin clusters.
Collapse
Affiliation(s)
- Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Cell and Molecular Biology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
49
|
Kumar A, Vadas O, Dos Santos Pacheco N, Zhang X, Chao K, Darvill N, Rasmussen HØ, Xu Y, Lin GMH, Stylianou FA, Pedersen JS, Rouse SL, Morgan ML, Soldati-Favre D, Matthews S. Structural and regulatory insights into the glideosome-associated connector from Toxoplasma gondii. eLife 2023; 12:e86049. [PMID: 37014051 PMCID: PMC10125020 DOI: 10.7554/elife.86049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/03/2023] [Indexed: 04/05/2023] Open
Abstract
The phylum of Apicomplexa groups intracellular parasites that employ substrate-dependent gliding motility to invade host cells, egress from the infected cells, and cross biological barriers. The glideosome-associated connector (GAC) is a conserved protein essential to this process. GAC facilitates the association of actin filaments with surface transmembrane adhesins and the efficient transmission of the force generated by myosin translocation of actin to the cell surface substrate. Here, we present the crystal structure of Toxoplasma gondii GAC and reveal a unique, supercoiled armadillo repeat region that adopts a closed ring conformation. Characterisation of the solution properties together with membrane and F-actin binding interfaces suggests that GAC adopts several conformations from closed to open and extended. A multi-conformational model for assembly and regulation of GAC within the glideosome is proposed.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Oscar Vadas
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Nicolas Dos Santos Pacheco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Xu Zhang
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Kin Chao
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Nicolas Darvill
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Helena Ø Rasmussen
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus UniversityAarhusDenmark
| | - Yingqi Xu
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Gloria Meng-Hsuan Lin
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | | | - Jan Skov Pedersen
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus UniversityAarhusDenmark
| | - Sarah L Rouse
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Marc L Morgan
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Stephen Matthews
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
50
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|