1
|
Jin X, Rosenbohm J, Moghaddam AO, Kim E, Seiffert-Sinha K, Leiker M, Zhai H, Baddam SR, Minnick G, Huo Y, Safa BT, Wahl JK, Meng F, Huang C, Lim JY, Conway DE, Sinha AA, Yang R. Desmosomal Cadherin Tension Loss in Pemphigus Vulgaris Mediated by the Inhibition of Active RhoA at Cell-Cell Adhesions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592394. [PMID: 38766211 PMCID: PMC11100601 DOI: 10.1101/2024.05.03.592394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Binding of autoantibodies to keratinocyte surface antigens, primarily desmoglein 3 (Dsg3) of the desmosomal complex, leads to the dissociation of cell-cell adhesion in the blistering disorder pemphigus vulgaris (PV). After the initial disassembly of desmosomes, cell-cell adhesions actively remodel in association with the cytoskeleton and focal adhesions. Growing evidence highlights the role of adhesion mechanics and mechanotransduction at cell-cell adhesions in this remodeling process, as their active participation may direct autoimmune pathogenicity. However, a large part of the biophysical transformations after antibody binding remains underexplored. Specifically, it is unclear how tension in desmosomes and cell-cell adhesions changes in response to antibodies, and how the altered tensional states translate to cellular responses. Here, we showed a tension loss at Dsg3 using fluorescence resonance energy transfer (FRET)-based tension sensors, a tension loss at the entire cell-cell adhesion, and a potentially compensatory increase in junctional traction force at cell-extracellular matrix adhesions after PV antibody binding. Further, our data indicate that this tension loss is mediated by the inhibition of RhoA at cell-cell contacts, and the extent of RhoA inhibition may be crucial in determining the severity of pathogenicity among different PV antibodies. More importantly, this tension loss can be partially restored by altering actomyosin based cell contractility. Collectively, these findings provide previously unattainable details in our understanding of the mechanisms that govern cell-cell interactions under physiological and autoimmune conditions, which may open the window to entirely new therapeutics aimed at restoring physiological balance to tension dynamics that regulates the maintenance of cell-cell adhesion.
Collapse
Affiliation(s)
- Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Amir Ostadi Moghaddam
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | | | - Merced Leiker
- Department of Dermatology, University at Buffalo, Buffalo, NY 14203
| | - Haiwei Zhai
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Sindora R. Baddam
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Yucheng Huo
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Republic of Singapore
| | - Bahareh Tajvidi Safa
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - James K. Wahl
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583
| | - Fanben Meng
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Changjin Huang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Republic of Singapore
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Daniel E. Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210
- The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210
| | - Animesh A. Sinha
- Department of Dermatology, University at Buffalo, Buffalo, NY 14203
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
2
|
Hariton WV, Schulze K, Rahimi S, Shojaeian T, Feldmeyer L, Schwob R, Overmiller AM, Sayar BS, Borradori L, Mahoney MG, Galichet A, Müller EJ. A desmosomal cadherin controls multipotent hair follicle stem cell quiescence and orchestrates regeneration through adhesion signaling. iScience 2023; 26:108568. [PMID: 38162019 PMCID: PMC10755723 DOI: 10.1016/j.isci.2023.108568] [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: 09/06/2023] [Revised: 11/03/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Stem cells (SCs) are critical to maintain tissue homeostasis. However, it is currently not known whether signaling through cell junctions protects quiescent epithelial SC reservoirs from depletion during disease-inflicted damage. Using the autoimmune model disease pemphigus vulgaris (PV), this study reveals an unprecedented role for a desmosomal cadherin in governing SC quiescence and regeneration through adhesion signaling in the multipotent mouse hair follicle compartment known as the bulge. Autoantibody-mediated, mechanical uncoupling of desmoglein (Dsg) 3 transadhesion activates quiescent bulge SC which lose their multipotency and stemness, become actively cycling, and finally delaminate from their epithelial niche. This then initiates a self-organized regenerative program which restores Dsg3 function and bulge morphology including SC quiescence and multipotency. These profound changes are triggered by the sole loss of functional Dsg3, resemble major signaling events in Dsg3-/- mice, and are driven by SC-relevant EGFR activation and Wnt modulation requiring longitudinal repression of Hedgehog signaling.
Collapse
Affiliation(s)
- William V.J. Hariton
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Katja Schulze
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Siavash Rahimi
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Taravat Shojaeian
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Laurence Feldmeyer
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Roman Schwob
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Andrew M. Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Beyza S. Sayar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Luca Borradori
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Mỹ G. Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Arnaud Galichet
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Eliane J. Müller
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
3
|
Perl AL, Koetsier JL, Green KJ. PP2A-B55alpha controls keratinocyte adhesion through dephosphorylation of the Desmoplakin C-terminus. Sci Rep 2023; 13:12720. [PMID: 37543698 PMCID: PMC10404246 DOI: 10.1038/s41598-023-37874-8] [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: 11/07/2022] [Accepted: 06/28/2023] [Indexed: 08/07/2023] Open
Abstract
Critical for the maintenance of epidermal integrity and function are attachments between intermediate filaments (IF) and intercellular junctions called desmosomes. The desmosomal cytoplasmic plaque protein desmoplakin (DP) is essential for anchoring IF to the junction. DP-IF interactions are regulated by a phospho-regulatory motif within the DP C-terminus controlling keratinocyte intercellular adhesion. Here we identify the protein phosphatase 2A (PP2A)-B55α holoenzyme as the major serine/threonine phosphatase regulating DP's C-terminus and consequent intercellular adhesion. Using a combination of chemical and genetic approaches, we show that the PP2A-B55α holoenzyme interacts with DP at intercellular membranes in 2D- and 3D- epidermal models and human skin samples. Our experiments demonstrate that PP2A-B55α regulates the phosphorylation status of junctional DP and is required for maintaining strong desmosome-mediated intercellular adhesion. These data identify PP2A-B55α as part of a regulatory module capable of tuning intercellular adhesion strength and a candidate disease target in desmosome-related disorders of the skin and heart.
Collapse
Affiliation(s)
- Abbey L Perl
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave., Chicago, IL, 60611, USA
| | - Jennifer L Koetsier
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave., Chicago, IL, 60611, USA
| | - Kathleen J Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave., Chicago, IL, 60611, USA.
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
4
|
Gonzalez JR, Celli A, Weckel A, Dhariwala MO, Merana GR, Ojewumi OT, Okoro J, Dwyer LR, Tran VM, Meyer JM, Mauro TM, Scharschmidt TC. FLG Deficiency in Mice Alters the Early-Life CD4 + T-Cell Response to Skin Commensal Bacteria. J Invest Dermatol 2022; 143:790-800.e12. [PMID: 36496196 DOI: 10.1016/j.jid.2022.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 12/12/2022]
Abstract
FLG variants underlie ichthyosis vulgaris and increased risk of atopic dermatitis, conditions typified by disruption of the skin microbiome and cutaneous immune response. Yet, it remains unclear whether neonatal skin barrier compromise because of FLG deficiency alters the quality of commensal-specific T cells and the functional impact of such responses. To address these questions, we profiled changes in the skin barrier and early cutaneous immune response of neonatal C57BL/6 Flg‒/‒ and wild-type mice using single-cell RNA sequencing, flow cytometry, and other modalities. Flg‒/‒ neonates showed little alteration in transepidermal water loss or lipid- or corneocyte-related gene expression. However, they showed increases in barrier disruption genes, epidermal dye penetration, and numbers of skin CD4+ T cells. Using an engineered strain of Staphylococcus epidermidis (S. epidermidis 2W) to study the response to neonatal skin colonization, we found that commensal-specific CD4+ T cells were skewed in Flg‒/‒ pups toward effector rather than regulatory T cells. This altered response persisted into adulthood, where it was typified by T helper 17 (Th17) cells and associated with increased susceptibility to imiquimod-induced skin inflammation. Thus, subtle but impactful differences in neonatal barrier function in Flg‒/‒ mice are accompanied by a skewed commensal-specific CD4+ response, with enduring consequences for skin immune homeostasis.
Collapse
Affiliation(s)
- Jeanmarie R Gonzalez
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, USA
| | - Anna Celli
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Dermatology Service, San Francisco VA Medical Center, San Francisco, California, USA
| | - Antonin Weckel
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Miqdad O Dhariwala
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Geil R Merana
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Oluwasunmisola T Ojewumi
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Joy Okoro
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Laura R Dwyer
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, USA
| | - Victoria M Tran
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, USA
| | - Jason M Meyer
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | - Theodora M Mauro
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Dermatology Service, San Francisco VA Medical Center, San Francisco, California, USA
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
5
|
Merecz-Sadowska A, Sitarek P, Zajdel K, Kucharska E, Kowalczyk T, Zajdel R. The Modulatory Influence of Plant-Derived Compounds on Human Keratinocyte Function. Int J Mol Sci 2021; 22:12488. [PMID: 34830374 PMCID: PMC8618348 DOI: 10.3390/ijms222212488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
The plant kingdom is a rich source of secondary metabolites with numerous properties, including the potential to modify keratinocyte biology. Keratinocytes are important epithelial cells that play a protective role against various chemical, physical and biological stimuli, and participate in reactive oxygen scavenging and inflammation and wound healing processes. The epidermal cell response may be modulated by phytochemicals via changes in signal transduction pathways. Plant extracts and single secondary compounds can possess a high antioxidant capacity and may suppress reactive oxygen species release, inhibit pro-apoptotic proteins and apoptosis and activate antioxidant enzymes in keratinocytes. Moreover, selected plant extracts and single compounds also exhibit anti-inflammatory properties and exposure may result in limited production of adhesion molecules, pro-inflammatory cytokines and chemokines in keratinocytes. In addition, plant extracts and single compounds may promote keratinocyte motility and proliferation via the regulation of growth factor production and enhance wound healing. While such plant compounds may modulate keratinocyte functions, further in vitro and in vivo studies are needed on their mechanisms of action, and more specific toxicity and clinical studies are needed to ensure their effectiveness and safety for use on human skin.
Collapse
Affiliation(s)
- Anna Merecz-Sadowska
- Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland;
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, 90-151 Lodz, Poland;
| | - Karolina Zajdel
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| | - Ewa Kucharska
- Chair of Gerontology, Geriatrics and Social Work at the Faculty of Pedagogy, Ignatianum Academy in Cracow, 31-501 Cracow, Poland;
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland;
| | - Radosław Zajdel
- Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland;
| |
Collapse
|
6
|
Paris S, Ekeanyanwu R, Jiang Y, Davis D, Spechler SJ, Souza RF. Obesity and its effects on the esophageal mucosal barrier. Am J Physiol Gastrointest Liver Physiol 2021; 321:G335-G343. [PMID: 34405732 DOI: 10.1152/ajpgi.00199.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity is associated with gastroesophageal reflux disease (GERD) and its complications including reflux esophagitis, Barrett's esophagus, and esophageal adenocarcinoma. Traditionally, these associations have been attributed to the mechanical effect of abdominal fat in increasing intra-abdominal pressure, thereby promoting gastroesophageal reflux and causing disruption of antireflux mechanisms at the esophagogastric junction. However, recent studies suggest that visceral adipose tissue (VAT) produces numerous cytokines that can cause esophageal inflammation and impair esophageal mucosal barrier integrity through reflux-independent mechanisms that render the esophageal mucosa especially susceptible to GERD-induced injury. In this report, we review mechanisms of esophageal mucosal defense, the genesis and remodeling of visceral adipose tissue during obesity, and the potential role of substances produced by VAT, especially the VAT that encircles the esophagogastric junction, in the impairment of esophageal mucosal barrier integrity that leads to the development of GERD complications.
Collapse
Affiliation(s)
- Shere Paris
- Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas
| | - Rebecca Ekeanyanwu
- Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Daniel Davis
- Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas.,Department of Surgery, Center for Esophageal Diseases, Baylor University Medical Center, Dallas, Texas
| | - Stuart Jon Spechler
- Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas.,Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center, Dallas, Texas
| | - Rhonda F Souza
- Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas.,Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center, Dallas, Texas
| |
Collapse
|
7
|
Cardiac Biomarkers and Autoantibodies in Endurance Athletes: Potential Similarities with Arrhythmogenic Cardiomyopathy Pathogenic Mechanisms. Int J Mol Sci 2021; 22:ijms22126500. [PMID: 34204386 PMCID: PMC8235133 DOI: 10.3390/ijms22126500] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
The “Extreme Exercise Hypothesis” states that when individuals perform training beyond the ideal exercise dose, a decline in the beneficial effects of physical activity occurs. This is due to significant changes in myocardial structure and function, such as hemodynamic alterations, cardiac chamber enlargement and hypertrophy, myocardial inflammation, oxidative stress, fibrosis, and conduction changes. In addition, an increased amount of circulating biomarkers of exercise-induced damage has been reported. Although these changes are often reversible, long-lasting cardiac damage may develop after years of intense physical exercise. Since several features of the athlete’s heart overlap with arrhythmogenic cardiomyopathy (ACM), the syndrome of “exercise-induced ACM” has been postulated. Thus, the distinction between ACM and the athlete’s heart may be challenging. Recently, an autoimmune mechanism has been discovered in ACM patients linked to their characteristic junctional impairment. Since cardiac junctions are similarly impaired by intense physical activity due to the strong myocardial stretching, we propose in the present work the novel hypothesis of an autoimmune response in endurance athletes. This investigation may deepen the knowledge about the pathological remodeling and relative activated mechanisms induced by intense endurance exercise, potentially improving the early recognition of whom is actually at risk.
Collapse
|
8
|
Broussard JA, Koetsier JL, Hegazy M, Green KJ. Desmosomes polarize and integrate chemical and mechanical signaling to govern epidermal tissue form and function. Curr Biol 2021; 31:3275-3291.e5. [PMID: 34107301 DOI: 10.1016/j.cub.2021.05.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/01/2021] [Accepted: 05/12/2021] [Indexed: 01/15/2023]
Abstract
The epidermis is a stratified epithelium in which structural and functional features are polarized across multiple cell layers. This type of polarity is essential for establishing the epidermal barrier, but how it is created and sustained is poorly understood. Previous work identified a role for the classic cadherin/filamentous-actin network in establishment of epidermal polarity. However, little is known about potential roles of the most prominent epidermal intercellular junction, the desmosome, in establishing epidermal polarity, in spite of the fact that desmosome constituents are patterned across the apical to basal cell layers. Here, we show that desmosomes and their associated intermediate filaments (IFs) are key regulators of mechanical polarization in epidermis, whereby basal and suprabasal cells experience different forces that drive layer-specific functions. Uncoupling desmosomes and IF or specific targeting of apical desmosomes through depletion of the superficial desmosomal cadherin, desmoglein 1, impedes basal stratification in an in vitro competition assay and suprabasal tight junction barrier functions in 3D reconstructed epidermis. Surprisingly, disengaging desmosomes from IF also accelerated the expression of differentiation markers, through precocious activation of the mechanosensitive transcriptional regulator serum response factor (SRF) and downstream activation of epidermal growth factor receptor family member ErbB2 by Src family kinase (SFK)-mediated phosphorylation. This Dsg1-SFK-ErbB2 axis also helps maintain tight junctions and barrier function later in differentiation. Together, these data demonstrate that the desmosome-IF network is a critical contributor to the cytoskeletal-adhesive machinery that supports the polarized function of the epidermis.
Collapse
Affiliation(s)
- Joshua A Broussard
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | | | - Marihan Hegazy
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA
| | - Kathleen J Green
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
9
|
Raya-Sandino A, Luissint AC, Kusters DHM, Narayanan V, Flemming S, Garcia-Hernandez V, Godsel LM, Green KJ, Hagen SJ, Conway DE, Parkos CA, Nusrat A. Regulation of intestinal epithelial intercellular adhesion and barrier function by desmosomal cadherin desmocollin-2. Mol Biol Cell 2021; 32:753-768. [PMID: 33596089 PMCID: PMC8108520 DOI: 10.1091/mbc.e20-12-0775] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/26/2022] Open
Abstract
The role of desmosomal cadherin desmocollin-2 (Dsc2) in regulating barrier function in intestinal epithelial cells (IECs) is not well understood. Here, we report the consequences of silencing Dsc2 on IEC barrier function in vivo using mice with inducible intestinal-epithelial-specific Dsc2 knockdown (KD) (Dsc2ERΔIEC). While the small intestinal gross architecture was maintained, loss of epithelial Dsc2 influenced desmosomal plaque structure, which was smaller in size and had increased intermembrane space between adjacent epithelial cells. Functional analysis revealed that loss of Dsc2 increased intestinal permeability in vivo, supporting a role for Dsc2 in the regulation of intestinal epithelial barrier function. These results were corroborated in model human IECs in which Dsc2 KD resulted in decreased cell-cell adhesion and impaired barrier function. It is noteworthy that Dsc2 KD cells exhibited delayed recruitment of desmoglein-2 (Dsg2) to the plasma membrane after calcium switch-induced intercellular junction reassembly, while E-cadherin accumulation was unaffected. Mechanistically, loss of Dsc2 increased desmoplakin (DP I/II) protein expression and promoted intermediate filament interaction with DP I/II and was associated with enhanced tension on desmosomes as measured by a Dsg2-tension sensor. In conclusion, we provide new insights on Dsc2 regulation of mechanical tension, adhesion, and barrier function in IECs.
Collapse
Affiliation(s)
- Arturo Raya-Sandino
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Dennis H. M. Kusters
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Sven Flemming
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | | | - Lisa M. Godsel
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kathleen J. Green
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Susan J. Hagen
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Daniel E. Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Charles A. Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
10
|
Jin X, Rosenbohm J, Kim E, Esfahani AM, Seiffert-Sinha K, Wahl JK, Lim JY, Sinha AA, Yang R. Modulation of Mechanical Stress Mitigates Anti-Dsg3 Antibody-Induced Dissociation of Cell-Cell Adhesion. Adv Biol (Weinh) 2021; 5:e2000159. [PMID: 33724731 PMCID: PMC7993752 DOI: 10.1002/adbi.202000159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/21/2020] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly clear that mechanical stress in adhesive junctions plays a significant role in dictating the fate of cell-cell attachment under physiological conditions. Targeted disruption of cell-cell junctions leads to multiple pathological conditions, among them the life-threatening autoimmune blistering disease pemphigus vulgaris (PV). The dissociation of cell-cell junctions by autoantibodies is the hallmark of PV, however, the detailed mechanisms that result in tissue destruction remain unclear. Thus far, research and therapy in PV have focused primarily on immune mechanisms upstream of autoantibody binding, while the biophysical aspects of the cell-cell dissociation process leading to acantholysis are less well studied. In work aimed at illuminating the cellular consequences of autoantibody attachment, it is reported that externally applied mechanical stress mitigates antibody-induced monolayer fragmentation and inhibits p38 MAPK phosphorylation activated by anti-Dsg3 antibody. Further, it is demonstrated that mechanical stress applied externally to cell monolayers enhances cell contractility via RhoA activation and promotes the strengthening of cortical actin, which ultimately mitigates antibody-induced cell-cell dissociation. The study elevates understanding of the mechanism of acantholysis in PV and shifts the paradigm of PV disease development from a focus solely on immune pathways to highlight the key role of physical transformations at the target cell.
Collapse
Affiliation(s)
- Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | | | - James K Wahl
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, 68583, USA
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Animesh A Sinha
- Department of Dermatology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
11
|
Karsch S, Büchau F, Magin TM, Janshoff A. An intact keratin network is crucial for mechanical integrity and barrier function in keratinocyte cell sheets. Cell Mol Life Sci 2020; 77:4397-4411. [PMID: 31912195 PMCID: PMC11104923 DOI: 10.1007/s00018-019-03424-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/25/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022]
Abstract
The isotype-specific composition of the keratin cytoskeleton is important for strong adhesion, force resilience, and barrier function of the epidermis. However, the mechanisms by which keratins regulate these functions are still incompletely understood. In this study, the role and significance of the keratin network for mechanical integrity, force transmission, and barrier formation were analyzed in murine keratinocytes. Following the time-course of single-cell wound closure, wild-type (WT) cells slowly closed the gap in a collective fashion involving tightly connected neighboring cells. In contrast, the mechanical response of neighboring cells was compromised in keratin-deficient cells, causing an increased wound area initially and an inefficient overall wound closure. Furthermore, the loss of the keratin network led to impaired, fragmented cell-cell junctions, and triggered a profound change in the overall cellular actomyosin architecture. Electric cell-substrate impedance sensing of cell junctions revealed a dysfunctional barrier in knockout (Kty-/-) cells compared to WT cells. These findings demonstrate that Kty-/- cells display a novel phenotype characterized by loss of mechanocoupling and failure to form a functional barrier. Re-expression of K5/K14 rescued the barrier defect to a significant extent and reestablished the mechanocoupling with remaining discrepancies likely due to the low abundance of keratins in that setting. Our study reveals the major role of the keratin network for mechanical homeostasis and barrier functionality in keratinocyte layers.
Collapse
Affiliation(s)
- Susanne Karsch
- Institute of Physical Chemistry, University of Göttingen, Göttingen, Germany
| | - Fanny Büchau
- Institute of Biology, University of Leipzig, Leipzig, Germany
| | - Thomas M Magin
- Institute of Biology, University of Leipzig, Leipzig, Germany.
| | - Andreas Janshoff
- Institute of Physical Chemistry, University of Göttingen, Göttingen, Germany.
| |
Collapse
|
12
|
Rübsam M, Broussard JA, Wickström SA, Nekrasova O, Green KJ, Niessen CM. Adherens Junctions and Desmosomes Coordinate Mechanics and Signaling to Orchestrate Tissue Morphogenesis and Function: An Evolutionary Perspective. Cold Spring Harb Perspect Biol 2018; 10:a029207. [PMID: 28893859 PMCID: PMC6211388 DOI: 10.1101/cshperspect.a029207] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cadherin-based adherens junctions (AJs) and desmosomes are crucial to couple intercellular adhesion to the actin or intermediate filament cytoskeletons, respectively. As such, these intercellular junctions are essential to provide not only integrity to epithelia and other tissues but also the mechanical machinery necessary to execute complex morphogenetic and homeostatic intercellular rearrangements. Moreover, these spatially defined junctions serve as signaling hubs that integrate mechanical and chemical pathways to coordinate tissue architecture with behavior. This review takes an evolutionary perspective on how the emergence of these two essential intercellular junctions at key points during the evolution of multicellular animals afforded metazoans with new opportunities to integrate adhesion, cytoskeletal dynamics, and signaling. We discuss known literature on cross-talk between the two junctions and, using the skin epidermis as an example, provide a model for how these two junctions function in concert to orchestrate tissue organization and function.
Collapse
Affiliation(s)
- Matthias Rübsam
- University of Cologne, Department of Dermatology, Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC) at the CECAD Research Center, 50931 Cologne, Germany
| | - Joshua A Broussard
- Northwestern University Feinberg School of Medicine, Departments of Pathology and Dermatology, the Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611
| | - Sara A Wickström
- Paul Gerson Unna Group, Skin Homeostasis and Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Oxana Nekrasova
- Northwestern University Feinberg School of Medicine, Departments of Pathology and Dermatology, the Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611
| | - Kathleen J Green
- Northwestern University Feinberg School of Medicine, Departments of Pathology and Dermatology, the Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611
| | - Carien M Niessen
- University of Cologne, Department of Dermatology, Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC) at the CECAD Research Center, 50931 Cologne, Germany
| |
Collapse
|
13
|
Sonavane PR, Wang C, Dzamba B, Weber GF, Periasamy A, DeSimone DW. Mechanical and signaling roles for keratin intermediate filaments in the assembly and morphogenesis of Xenopus mesendoderm tissue at gastrulation. Development 2017; 144:4363-4376. [PMID: 28982683 PMCID: PMC5769636 DOI: 10.1242/dev.155200] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/25/2017] [Indexed: 12/21/2022]
Abstract
The coordination of individual cell behaviors is a crucial step in the assembly and morphogenesis of tissues. Xenopus mesendoderm cells migrate collectively along a fibronectin (FN) substrate at gastrulation, but how the adhesive and mechanical forces required for these movements are generated and transmitted is unclear. Traction force microscopy (TFM) was used to establish that traction stresses are limited primarily to leading edge cells in mesendoderm explants, and that these forces are balanced by intercellular stresses in follower rows. This is further reflected in the morphology of these cells, with broad lamellipodial protrusions, mature focal adhesions and a gradient of activated Rac1 evident at the leading edge, while small protrusions, rapid turnover of immature focal adhesions and lack of a Rac1 activity gradient characterize cells in following rows. Depletion of keratin (krt8) with antisense morpholinos results in high traction stresses in follower row cells, misdirected protrusions and the formation of actin stress fibers anchored in streak-like focal adhesions. We propose that maintenance of mechanical integrity in the mesendoderm by keratin intermediate filaments is required to balance stresses within the tissue to regulate collective cell movements.
Collapse
Affiliation(s)
- Pooja R Sonavane
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Chong Wang
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Bette Dzamba
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Gregory F Weber
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Ammasi Periasamy
- Keck Center for Cellular Imaging, Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Douglas W DeSimone
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| |
Collapse
|
14
|
Yu C, Li Y, Liu M, Gao M, Li C, Yan H, Li C, Sun L, Mo L, Wu C, Qi X, Ren J. Critical Role of Hepatic Cyp450s in the Testis-Specific Toxicity of (5R)-5-Hydroxytriptolide in C57BL/6 Mice. Front Pharmacol 2017; 8:832. [PMID: 29209210 PMCID: PMC5702336 DOI: 10.3389/fphar.2017.00832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/31/2017] [Indexed: 12/17/2022] Open
Abstract
Low solubility, tissue accumulation, and toxicity are chief obstacles to developing triptolide derivatives, so a better understanding of the pharmacokinetics and toxicity of triptolide derivatives will help with these limitations. To address this, we studied pharmacokinetics and toxicity of (5R)-5-hydroxytriptolide (LLDT-8), a novel triptolide derivative immunosuppressant in a conditional knockout (KO) mouse model with liver-specific deletion of CYP450 reductase. Compared to wild type (WT) mice, after LLDT-8 treatment, KO mice suffered severe testicular toxicity (decreased testicular weight, spermatocytes apoptosis) unlike WT mice. Moreover, KO mice had greater LLDT-8 exposure as confirmed with elevated AUC and Cmax, increased drug half-life, and greater tissue distribution. γ-H2AX, a marker of meiosis process, its localization and protein level in testis showed a distinct meiosis block induced by LLDT-8. RNA polymerase II (Pol II), an essential factor for RNA storage and synapsis in spermatogenesis, decreased in testes of KO mice after LLDT-8 treatment. Germ-cell line based assays confirmed that LLDT-8 selectively inhibited Pol II in spermatocyte-like cells. Importantly, the analysis of androgen receptor (AR) related genes showed that LLDT-8 did not change AR-related signaling in testes. Thus, hepatic CYP450s were responsible for in vivo metabolism and clearance of LLDT-8 and aggravated testicular injury may be due to increased LLDT-8 exposure in testis and subsequent Pol II reduction.
Collapse
Affiliation(s)
- Cunzhi Yu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yu Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mingxia Liu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Man Gao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chenggang Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hong Yan
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunzhu Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lihan Sun
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Liying Mo
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Lv Y, Fu L. The potential mechanism for Hydroxysafflor yellow A attenuating blood-brain barrier dysfunction via tight junction signaling pathways excavated by an integrated serial affinity chromatography and shotgun proteomics analysis approach. Neurochem Int 2017; 112:38-48. [PMID: 29107696 DOI: 10.1016/j.neuint.2017.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 12/15/2022]
Abstract
Our previous studies elucidated that hydroxysafflor yellow A (HSYA) exerted anti-inflammatory effects against ischemia stroke by inhibiting TLR4 pathway-mediated signaling transduction. However, only several targets were verified in that limited work. The integrated method of serial affinity chromatography (SAC) and shotgun proteomics analysis (SPA) might be an alternative approach for exploring a potential therapeutic role. SAC was induced to extract specific binding proteins in the brain tissue of 2 h of ischemia stroke mice via HSYA affinity matrices. SPA was conducted by nanoLC-MS/MS, while the identified proteins were mapped on to Gene Ontology and KEGG pathway components analysis. The protection of HSYA for blood-brain barrier in mice with ischemia stroke was assessed with the leakage of Evans Blue. The expression of tight junction proteins of blood-brain barrier: occludin, claudin-5, and ZO-1 were detected with ischemia boundary positive areas staining. The regulation of nonmuscle myosin heavy chain IIA (NMMHC IIA), TLR4-mediated PI3K/AKT/JNK1/2/14-3-3ε/NF-κB p65 signaling pathway were evaluated using western blot analysis. A total of 35 proteins with molecular eights ranging from 27,841.22 to 234,122.79 KD were identified. Gene Ontology annotation and KEGG pathways analysis of the identified proteins were conducted with tight junction and PI3K/AKT signaling pathways. HSYA could significantly reduce the leakage of Evans Blue in mice with ischemia stroke, while attenuating the expression of occludin, claudin-5, and ZO-1. Western blot demonstrated that regulation of NMMHC IIA, TLR4-mediated PI3K/AKT/JNK1/2/14-3-3ε/NF-κB p65 signaling pathway played an essential role in the protective effect of HSYA. The integrated method of SAC and SPA provides the promising explanations for exploring the mechanism underlying blood-brain barrier dysfunction via the tight junction pathway. HSYA could attenuate blood-brain barrier dysfunction in anti-inflammatory patterns in ischemia stroke mice via the tight junction pathway.
Collapse
Affiliation(s)
- Yanni Lv
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Longsheng Fu
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
16
|
Broussard JA, Yang R, Huang C, Nathamgari SSP, Beese AM, Godsel LM, Hegazy MH, Lee S, Zhou F, Sniadecki NJ, Green KJ, Espinosa HD. The desmoplakin-intermediate filament linkage regulates cell mechanics. Mol Biol Cell 2017; 28:3156-3164. [PMID: 28495795 PMCID: PMC5687018 DOI: 10.1091/mbc.e16-07-0520] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 03/16/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
Desmoplakin connects desmosomal core components to intermediate filaments at sites of cell–cell adhesion. Modulating the strength of this linkage using desmoplakin mutants led to alterations in cell–substrate and cell–cell forces and cell stiffness as assessed by micropillar arrays and atomic force microscopy. Perturbation of the actin cytoskeleton leads to abrogation of these effects. The translation of mechanical forces into biochemical signals plays a central role in guiding normal physiological processes during tissue development and homeostasis. Interfering with this process contributes to cardiovascular disease, cancer progression, and inherited disorders. The actin-based cytoskeleton and its associated adherens junctions are well-established contributors to mechanosensing and transduction machinery; however, the role of the desmosome–intermediate filament (DSM–IF) network is poorly understood in this context. Because a force balance among different cytoskeletal systems is important to maintain normal tissue function, knowing the relative contributions of these structurally integrated systems to cell mechanics is critical. Here we modulated the interaction between DSMs and IFs using mutant forms of desmoplakin, the protein bridging these structures. Using micropillar arrays and atomic force microscopy, we demonstrate that strengthening the DSM–IF interaction increases cell–substrate and cell–cell forces and cell stiffness both in cell pairs and sheets of cells. In contrast, disrupting the interaction leads to a decrease in these forces. These alterations in cell mechanics are abrogated when the actin cytoskeleton is dismantled. These data suggest that the tissue-specific variability in DSM–IF network composition provides an opportunity to differentially regulate tissue mechanics by balancing and tuning forces among cytoskeletal systems.
Collapse
Affiliation(s)
- Joshua A Broussard
- Department of Pathology, Northwestern University, Chicago, IL 60611.,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Ruiguo Yang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Changjin Huang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - S Shiva P Nathamgari
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Allison M Beese
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Lisa M Godsel
- Department of Pathology, Northwestern University, Chicago, IL 60611.,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Marihan H Hegazy
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Sherry Lee
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Fan Zhou
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195.,Department of Bioengineering, University of Washington, Seattle, WA 98195.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195
| | - Kathleen J Green
- Department of Pathology, Northwestern University, Chicago, IL 60611 .,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208 .,Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL 60208
| |
Collapse
|
17
|
Markov AG, Aschenbach JR, Amasheh S. The epithelial barrier and beyond: Claudins as amplifiers of physiological organ functions. IUBMB Life 2017; 69:290-296. [PMID: 28371008 DOI: 10.1002/iub.1622] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/03/2017] [Indexed: 12/22/2022]
Abstract
Epithelial cell layers are interconnected by a meshwork of tight junction (TJ) protein strands, which are localized within apicolateral membranes. The proteins that form TJs are regarded to provide a static barrier, determining epithelial properties. However, recent findings in the field of barriology suggest that TJs contribute to more physiological aspects than indicated by the sum of the qualities of the single TJ proteins. Generally, TJs exhibit four major functions: (i) a "gate function," defining transepithelial permeability (i.e., barrier) properties, (ii) a "fence function" determining epithelial cell polarity, (iii) a "signaling function," affecting regulatory pathways, and (iv) a "stabilizing function," maintaining the integrity of the epithelium. This review presents a critical view on how the efficacy of physiological processes in epithelia and thus organ function might be improved by changes in the expression of claudins, the latter representing the largest and most variable family of TJ proteins. Major focus is set on (i) the coordinated regulation of transport and barrier in the intestine, (ii) the role of TJs in defining the route for antigen uptake and presentation in intestinal Peyer's patches, and (iii) the TJ function in mammary glands in response to milk accumulation, which represent impressive examples to highlight the amplification of epithelial functions by TJ proteins. © 2017 IUBMB Life, 69(5):290-296, 2017.
Collapse
Affiliation(s)
- Alexander G Markov
- Department of General Physiology, St. Petersburg State University, Russia
| | - Jörg R Aschenbach
- Department of Veterinary Medicine, Freie Universität Berlin, Institute of Veterinary Physiology, Berlin, Germany
| | - Salah Amasheh
- Department of Veterinary Medicine, Freie Universität Berlin, Institute of Veterinary Physiology, Berlin, Germany
| |
Collapse
|
18
|
Di Zenzo G, Amber KT, Sayar BS, Müller EJ, Borradori L. Immune response in pemphigus and beyond: progresses and emerging concepts. Semin Immunopathol 2015; 38:57-74. [DOI: 10.1007/s00281-015-0541-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/30/2015] [Indexed: 12/18/2022]
|
19
|
Abstract
Cell-cell adhesions are necessary for structural integrity and barrier formation of the epidermis. Here, we discuss insights from genetic and cell biological studies into the roles of individual cell-cell junctions and their composite proteins in regulating epidermal development and function. In addition to individual adhesive functions, we will discuss emerging ideas on mechanosensation/transduction of junctions in the epidermis, noncanonical roles for adhesion proteins, and crosstalk/interdependencies between the junctional systems. These studies have revealed that cell adhesion proteins are connected to many aspects of tissue physiology including growth control, differentiation, and inflammation.
Collapse
Affiliation(s)
- Kaelyn D Sumigray
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Terry Lechler
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
20
|
Patel DM, Dubash AD, Kreitzer G, Green KJ. Disease mutations in desmoplakin inhibit Cx43 membrane targeting mediated by desmoplakin-EB1 interactions. ACTA ACUST UNITED AC 2014; 206:779-97. [PMID: 25225338 PMCID: PMC4164953 DOI: 10.1083/jcb.201312110] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mechanisms by which microtubule plus ends interact with regions of cell-cell contact during tissue development and morphogenesis are not fully understood. We characterize a previously unreported interaction between the microtubule binding protein end-binding 1 (EB1) and the desmosomal protein desmoplakin (DP), and demonstrate that DP-EB1 interactions enable DP to modify microtubule organization and dynamics near sites of cell-cell contact. EB1 interacts with a region of the DP N terminus containing a hotspot for pathogenic mutations associated with arrhythmogenic cardiomyopathy (AC). We show that a subset of AC mutations, in addition to a mutation associated with skin fragility/woolly hair syndrome, impair gap junction localization and function by misregulating DP-EB1 interactions and altering microtubule dynamics. This work identifies a novel function for a desmosomal protein in regulating microtubules that affect membrane targeting of gap junction components, and elucidates a mechanism by which DP mutations may contribute to the development of cardiac and cutaneous diseases.
Collapse
Affiliation(s)
- Dipal M Patel
- Department of Pathology and Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Adi D Dubash
- Department of Pathology and Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Geri Kreitzer
- Department of Cell and Developmental Biology, Weill Cornell Medical College of Cornell University, New York, NY 10065
| | - Kathleen J Green
- Department of Pathology and Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 Department of Pathology and Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|