1
|
Papafilippou E, Baldauf L, Charras G, Kabla AJ, Bonfanti A. Interplay of damage and repair in the control of epithelial tissue integrity in response to cyclic loading. Curr Opin Cell Biol 2025; 94:102511. [PMID: 40233605 DOI: 10.1016/j.ceb.2025.102511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
Epithelial tissues are continuously exposed to cyclic stretch in vivo. Physiological stretching has been found to regulate soft tissue function at the molecular, cellular, and tissue scales, allowing tissues to preserve their homeostasis and adapt to challenges. In contrast, dysregulated or pathological stretching can induce damage and tissue fragilisation. Many mechanisms have been described for the repair of epithelial tissues across a range of timescales. In this review, we present the timescales of (i) physiological cyclic loading regimes, (ii) strain-regulated remodeling and damage accumulation, and (iii) repair mechanisms in epithelial tissues. We discuss how the response to cyclic loading in biological tissues differs from synthetic materials, in that damage can be partially or fully reversed by repair mechanisms acting on timescales shorter than cyclic loading. We highlight that timescales are critical to understanding the interplay between damage and repair in tissues that experience cyclic loading, opening up new avenues for exploring soft tissue homeostasis.
Collapse
Affiliation(s)
| | - Lucia Baldauf
- London Centre for Nanotechnology, University College London, London, UK
| | - Guillaume Charras
- London Centre for Nanotechnology, University College London, London, UK; Institute for the Physics of Living Systems, University College London, London, UK; Department of Cell and Developmental Biology, University College London, London, UK.
| | | | - Alessandra Bonfanti
- Department of Civil and Environmental Engineering, Politecnico di Milano, Milan, Italy.
| |
Collapse
|
2
|
Lee SJ, Wu Z, Huang M, Liang C, Huang Z, Chen S, Rajasekar V, Abdalla MM, Nah H, Heo DN, Kwon IK, Cho MJ, Kim SJ, Sohn S, Kim SH, Sugimura R, Yiu CKY. Crosslinker-free in situ hydrogel induces self-aggregation of human dental pulp stem cells with enhanced antibacterial activity. Mater Today Bio 2025; 31:101451. [PMID: 39896283 PMCID: PMC11783010 DOI: 10.1016/j.mtbio.2025.101451] [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: 08/24/2024] [Revised: 10/29/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Recently, injectable hydrogels have garnered significant attention in tissue engineering due to their controlled flowability, strong plasticity, adaptability, and good biocompatibility. However, research on readily injectable in situ-forming hydrogels capable of forming functional three-dimensional (3D) tissue condensations remains limited. This study explores the development and evaluation of a carboxymethyl chitosan (CMCTS)/oxidized hyaluronic acid (oHA) hydrogel incorporated with silver sulfadiazine (AgSD) for tissue engineering applications with inherent antibacterial activity. Through physicochemical analysis, the optimal formulation of CMCTS/oHA hydrogels was established. The hydrogel demonstrated excellent injectability, enabling minimally invasive in situ delivery. In vitro cytotoxicity assays identified 0.1 % AgSD as the optimal concentration, supporting cell proliferation while exhibiting antimicrobial efficacy against S. mutans and E. faecalis. In vivo studies revealed complete hydrogel degradation and good biocompatibility, with no adverse tissue reactions. The hydrogel's ability to form 3D cell aggregates and support tissue regeneration underscores its potential for future 3D tissue engineering applications. Consequently, the injectable CMCTS/oHA/AgSD hydrogel developed in this study holds significant potential for application in a wide range of bioengineering fields, including antibacterial substance delivery systems and 3D tissue engineering, indicating potential for future clinical application.
Collapse
Affiliation(s)
- Sang Jin Lee
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Zhenzhen Wu
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Mengyu Huang
- Paediatric Dentistry, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Chao Liang
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Ziqi Huang
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Siyuan Chen
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Vidhyashree Rajasekar
- Paediatric Dentistry, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Mohamed Mahmoud Abdalla
- Restorative Dental Sciences, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong, China
- Dental Biomaterials Department, Faculty of Dental Medicine, Al-Azhar University, Cairo, Egypt
| | - Haram Nah
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Dong Nyoung Heo
- Biofriends Inc, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, South Korea
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Min-Jai Cho
- Department of Neurosurgery, Chungbuk National University College of Medicine, Chungbuk National University Hospital, Seowon-gu, Cheongju-si, 28644, Chungcheong-do, Republic of Korea
| | - Seong Jun Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan, 49315, Republic of Korea
| | - Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
- Centre for Translational Stem Cell Biology, Hong Kong SAR, PR China
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| |
Collapse
|
3
|
Schoon RM, van der Meer WJ, van Stalborch AMD, van Buul JD, Huveneers S. VE-cadherin RGD motifs are dispensable for cell-cell junctions, endothelial barrier function and monocyte extravasation. Tissue Barriers 2025:2478349. [PMID: 40099517 DOI: 10.1080/21688370.2025.2478349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
VE-cadherin is a key transmembrane protein involved in endothelial cell-cell junctions, playing a crucial role in maintaining vascular integrity and regulating selective leukocyte extravasation into inflamed tissue. The extracellular domain of human VE-cadherin contains two arginine-glycine-aspartate (RGD) motifs, which are known integrin-binding sites within extracellular matrix proteins, particularly for integrins of the β1, β3, and β5 families. In this study, we examined the functional relevance of these RGD motifs by generating VE-cadherin variants in which the RGD sequences were mutated to nonfunctional RGE. Immunofluorescence analysis showed that the VE-cadherin [D238E], VE-cadherin [D301E], and double-mutant VE-cadherin [D238/301E] variants formed stable endothelial cell-cell junctions that were comparable to junctions based on wild-type VE-cadherin. Additionally, electric cell-substrate impedance sensing (ECIS) confirmed that endothelial cells expressing each VE-cadherin RGD>RGE variant maintained efficient barrier function capacity. Moreover, monocyte transmigration assays demonstrated that the RGD>RGE mutations did not affect monocyte-endothelial interactions during transmigration. In summary, our findings indicate that the VE-cadherin RGD motifs are not essential for endothelial junction formation or monocyte transmigration.
Collapse
Affiliation(s)
- Rianne M Schoon
- Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Jaap D van Buul
- Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
4
|
Feng P, He C, Li G, Li J, Luo Y, Chen Y, Tang Y, Ma J, Ke C. A light-cured injectable composite hydrogel based on chitosan and decellularized matrix modulates stem cell aggregation behavior for accelerating cartilage defect repair. Int J Biol Macromol 2025; 295:139711. [PMID: 39798753 DOI: 10.1016/j.ijbiomac.2025.139711] [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: 08/23/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Cartilage repair remains a formidable challenge because of its limited regenerative capacity. Construction of a biomimetic hydrogel matrix that can induce cell aggregation is a promising therapeutic option. Cell aggregates are more beneficial than dissociated cells for improving survival and chondrogenic differentiation, thereby facilitating cartilage repair. Herein, we report a light-cured injectable composite hydrogel with cellular aggregation properties for cartilage defect repair that combines methacrylated chitosan (CSMA) and a methacrylate-modified decellularized extracellular matrix (dECMMA). The CSMA promotes cell aggregate formation by enhancing cadherin expression. The dECMMA retains many inherent components and bioactive factors, thereby providing a more natural microenvironment for cell proliferation and differentiation. By precisely adjusting the compositions of the CSMA and dECMMA, it was possible to fine-tune their physicochemical properties and biochemical cues. The results showed that a composite hydrogel composed of 2 % (m/v) CSMA and 5 % (m/v) dECMMA exhibited good injectability, appropriate degradability and mechanical properties, and regulated the formation of bone marrow mesenchymal stem cell aggregates. Transplantation of the composite hydrogel loaded with bone marrow mesenchymal stem cells (BMSCs) into a cartilage defect model demonstrated effective hyaline cartilage repair. Thus, light-cured injectable composite hydrogels embedded with BMSCs holds clinical promise for cartilage defect repair.
Collapse
Affiliation(s)
- Peipei Feng
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Chaonan He
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Guanrong Li
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Jin Li
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Yang Luo
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yaoqi Chen
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yun Tang
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China
| | - Jingyun Ma
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China.
| | - Chunhai Ke
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China.
| |
Collapse
|
5
|
Troyanovsky RB, Indra I, Troyanovsky SM. Actin-dependent α-catenin oligomerization contributes to adherens junction assembly. Nat Commun 2025; 16:1801. [PMID: 39979305 PMCID: PMC11842732 DOI: 10.1038/s41467-025-57079-z] [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] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Classic cadherins, specifically E-cadherin in most epithelial cells, are transmembrane adhesion receptors, whose intracellular region interacts with proteins, termed catenins, forming the cadherin-catenin complex (CCC). The cadherin ectodomain generates 2D adhesive clusters (E-clusters) through cooperative trans and cis interactions, while catenins anchor the E-clusters to the actin cytoskeleton. How these two types of interactions are coordinated in the formation of specialized cell-cell adhesions, adherens junctions (AJ), remains unclear. Here, we focus on the role of the actin-binding domain of α-catenin (αABD) by showing that the interaction of the αABD with actin generates actin-bound linear CCC oligomers (CCC/actin strands) incorporating up to six CCCs. This actin-driven CCC oligomerization, which is cadherin ectodomain independent, preferentially occurs along the actin cortex enriched with key basolateral proteins, myosin-1c, scribble, and DLG1. In cell-cell contacts, the CCC/actin strands integrate with the E-clusters giving rise to the composite oligomers, E/actin clusters. Targeted inactivation of strand formation by point mutations emphasizes the importance of this oligomerization process for blocking intercellular protrusive membrane activity and for coupling AJs with the actomyosin-derived tensional forces.
Collapse
Affiliation(s)
- Regina B Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Indrajyoti Indra
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Department of Cell & Developmental Biology, The Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
Choi W, Goldfarb D, Yan F, Major MB, Fanning AS, Peifer M. Proximity proteomics provides a new resource for exploring the function of Afadin and the complexity of cell-cell adherens junctions. Biol Open 2025; 14:bio061811. [PMID: 39882731 PMCID: PMC11810119 DOI: 10.1242/bio.061811] [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: 11/07/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
The network of proteins at the interface between cell-cell adherens junctions and the actomyosin cytoskeleton provides robust yet dynamic connections that facilitate cell shape change and motility. While this was initially thought to be a simple linear connection via classic cadherins and their associated catenins, we now have come to appreciate that many more proteins are involved, providing robustness and mechanosensitivity. Defining the full set of proteins in this network remains a key objective in our field. Proximity proteomics provides a means to define these networks. Mammalian Afadin and its Drosophila homolog Canoe are key parts of this protein network, facilitating diverse cell shape changes during gastrulation and other events of embryonic morphogenesis. Here we report results of several proximity proteomics screens, defining proteins in the neighborhood of both the N- and C-termini of mammalian Afadin in the premier epithelial model, MDCK cells. We compare our results with previous screens done in other cell types, and with proximity proteomics efforts with other junctional proteins. These reveal the value of multiple screens in defining the full network of neighbors and offer interesting insights into the overlap in protein composition between different epithelial cell junctions.
Collapse
Affiliation(s)
- Wangsun Choi
- Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA
| | - Dennis Goldfarb
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA63110
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine, St. Louis, MO, USA63110
| | - Feng Yan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michael B. Major
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA63110
| | - Alan S. Fanning
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark Peifer
- Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
7
|
Gong R, Reynolds MJ, Sun X, Alushin GM. Afadin mediates cadherin-catenin complex clustering on F-actin linked to cooperative binding and filament curvature. SCIENCE ADVANCES 2025; 11:eadu0989. [PMID: 39951520 PMCID: PMC11827635 DOI: 10.1126/sciadv.adu0989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
The E-cadherin-β-catenin-αE-catenin (cadherin-catenin) complex couples the cytoskeletons of neighboring cells at adherens junctions (AJs) to mediate force transmission across epithelia. Mechanical force and auxiliary binding partners converge to stabilize the cadherin-catenin complex's inherently weak binding to actin filaments (F-actin) through unclear mechanisms. Here, we show that afadin's coiled-coil (CC) domain and vinculin synergistically enhance the cadherin-catenin complex's F-actin engagement. The cryo-electron microscopy (cryo-EM) structure of an E-cadherin-β-catenin-αE-catenin-vinculin-afadin-CC supra-complex bound to F-actin reveals that afadin-CC bridges adjacent αE-catenin actin-binding domains along the filament, stabilizing flexible αE-catenin segments implicated in mechanical regulation. These cooperative binding contacts promote the formation of supra-complex clusters along F-actin. Additionally, cryo-EM variability analysis links supra-complex binding along individual F-actin strands to nanoscale filament curvature, a deformation mode associated with cytoskeletal forces. Collectively, this work elucidates a mechanistic framework by which vinculin and afadin tune cadherin-catenin complex-cytoskeleton coupling to support AJ function across varying mechanical regimes.
Collapse
Affiliation(s)
- Rui Gong
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Matthew J. Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Gregory M. Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
8
|
Wang J, Zhang G, Xing K, Wang B, Liu Y, Xue Y, Liu S, Leong DT. Influencing inter-cellular junctions with nanomaterials. Adv Colloid Interface Sci 2025; 336:103372. [PMID: 39671889 DOI: 10.1016/j.cis.2024.103372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/15/2024]
Abstract
Cell-cell junctions are essential for maintaining tissue integrity and regulating a wide range of physiological processes. While the disruption of intercellular junctions may lead to pathological conditions, it also presents an opportunity for therapeutic interventions. Nanomaterials have emerged as promising tools for modulating cell-cell junctions, offering new avenues for innovative treatments. In this review, we provide a comprehensive overview of the various nanomaterials interaction with cell-cell junctions. We discussed their underlying mechanisms, heterogenous effects on cellular behavior, and the therapeutic strategies of applying nanomaterial-induced intercellular junction disruption. Additionally, we address the challenges and opportunities involved in translating these strategies into clinical practice and discuss future directions for this rapidly advancing field.
Collapse
Affiliation(s)
- Jinping Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | - Guoying Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kuoran Xing
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | - Baoteng Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yanping Liu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Yuling Xue
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Shankui Liu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| |
Collapse
|
9
|
Yu J, Mabrouk I, Liu Q, Zhou Y, Song Y, Ma J, Liu F, Hu X, Yang Z, Zeng Y, Hu J, Sun Y. Transcriptomic analysis of melatonin on the mechanism of embryonic gonadal development in female Jilin white geese. Poult Sci 2025; 104:104527. [PMID: 39566171 PMCID: PMC11617457 DOI: 10.1016/j.psj.2024.104527] [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: 09/08/2024] [Revised: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
The goose industry represents a significant sector within the broader waterfowl industry in China, with early gonadal development playing a pivotal role in enhancing population productivity. Melatonin plays a crucial role in regulating early gonadal development, but the molecular mechanism underlying the effect of melatonin as a regulator remains unclear. In this study, the relationship between melatonin and egg production performance in Jilin white goose was first established, then, an in Ovo injection of melatonin in goose embryos on the embryonic day 12 (E12) was performed to evaluate the impact of melatonin on the gonadal development in Jilin white goose. The results showed that the in Ovo injection of melatonin increased the expression of FOXL2 in the gonads while decreasing the expression of CYP19A1. Transcriptome analyses revealed that a total of 1184 differentially expressed genes were screened in Jilin white goose embryonic gonads among the two comparison groups. These DEGs were enriched in Gene Ontology (GO) terms related to the microtubule cytoskeleton, cytoskeleton, cell adhesion, and biological adhesion and the KEGG enrichment analysis suggested that the DEGs were mainly enriched in "MAPK signaling pathway", "Wnt signaling pathway", "Calcium signaling pathway", "Focal adhesion", "Neuroactive ligand-receptor interaction", "Melanogenesis", "mTOR signaling pathway", "ECM-receptor interaction", and "TGF-beta signaling pathway" in the two comparison groups. The protein-protein interaction network analysis (PPI) indicated that selected DEGs, such as PDIA3, ATCB, PKM, PGAM1, NME2, LDHB, and CALM2, were highly related to the regulation of the reproduction system development. Additionally, the expression trends of 4 identified DEGs were validated by RT-qPCR. In conclusion, this study elucidates the potential regulatory role of melatonin in gonadal development in geese, identifying candidate genes involved in early gonadal differentiation and maturation. Furthermore, the findings offer valuable practical implications for the goose industry, providing professionals with scientifically grounded strategies to enhance reproductive efficiency and overall population productivity.
Collapse
Affiliation(s)
- Jin Yu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Ichraf Mabrouk
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qiuyuan Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yuxuan Zhou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yupu Song
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jingyun Ma
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Fengshuo Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xiangman Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Zhiyi Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yuxia Zeng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jingtao Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yongfeng Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Jilin Agricultural University, Ministry of Education, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
10
|
Gandhi AK, Huang YH, Sun ZYJ, Kim WM, Kondo Y, Hanley T, Beauchemin N, Blumberg RS. Structural aspects of CEACAM1 interactions. Eur J Clin Invest 2024; 54 Suppl 2:e14357. [PMID: 39555955 DOI: 10.1111/eci.14357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a membrane protein that plays an important role in a variety of immune and non-immune functions. Such functions are regulated by its activity as a homophilic ligand but also through its ability to interact as a heterophilic ligand with various host proteins. These include CEACAM5, T cell immunoglobulin-mucin like protein-3 (TIM-3) and, potentially, protein death protein 1 (PD-1). Furthermore, CEACAM1 is targeted by various pathogens to allow them to invade a host and bypass an effective immune response. Clinically, CEACAM1 plays an important role in infectious diseases, autoimmunity and cancer. In this review, we describe the structural basis for CEACAM1 interactions as a homophilic and heterophilic ligand. We discuss the regulation of its monomeric, dimeric and oligomeric states in cis and trans binding as well as the consequences for eliciting downstream signalling activities. Furthermore, we explore the potential role of avidity in determining CEACAM1's activities.
Collapse
Affiliation(s)
- Amit K Gandhi
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yu-Hwa Huang
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhen-Yu J Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Walter M Kim
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yasuyuki Kondo
- Division of Gastroenterology, Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Thomas Hanley
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Canada
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Xiong W, Shu XL, Huang L, He SQ, Liu LH, Li S, Shao ZC, Wang J, Cheng L. Bioinformatics Analysis and Experimental Validation of Differential Genes and Pathways in Bone Nonunions. Biochem Genet 2024; 62:4494-4517. [PMID: 38324134 DOI: 10.1007/s10528-023-10633-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/12/2023] [Indexed: 02/08/2024]
Abstract
Non-union fractures pose a significant clinical challenge, often leading to prolonged pain and disability. Understanding the molecular mechanisms underlying non-union fractures is crucial for developing effective therapeutic interventions. This study integrates bioinformatics analysis and experimental validation to unravel key genes and pathways associated with non-union fractures. We identified differentially expressed genes (DEGs) between non-union and fracture healing tissues using bioinformatics techniques. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed to elucidate the biological processes and pathways involved. Common DEGs were identified, and a protein-protein interaction (PPI) network was constructed. Fibronectin-1 (FN1), Thrombospondin-1 (THBS1), and Biglycan (BGN) were pinpointed as critical target genes for non-union fracture treatment. Experimental validation involved alkaline phosphatase (ALP) and Alizarin Red staining to confirm osteogenic differentiation. Our analysis revealed significant alterations in pathways related to cell behavior, tissue regeneration, wound healing, infection, and immune responses in non-union fracture tissues. FN1, THBS1, and BGN were identified as key genes, with their upregulation indicating potential disruptions in the bone remodeling process. Experimental validation confirmed the induction of osteogenic differentiation. The study provides comprehensive insights into the molecular mechanisms of non-union fractures, emphasizing the pivotal roles of FN1, THBS1, and BGN in extracellular matrix dynamics and bone regeneration. The findings highlight potential therapeutic targets and pathways for further investigation. Future research should explore interactions between these genes, validate results using in vivo fracture models, and develop tailored treatment strategies for non-union fractures, promising significant advances in clinical management.
Collapse
Affiliation(s)
- Wei Xiong
- Rehabilitation Medicine Department, Nanchang Hongdu Hospital of Traditional Chinese Medicine, No. 264, Minde Road, Donghu District, Nanchang City, 330008, Jiangxi, China
| | - Xing-Li Shu
- Rehabilitation Medicine Department, Nanchang Hongdu Hospital of Traditional Chinese Medicine, No. 264, Minde Road, Donghu District, Nanchang City, 330008, Jiangxi, China
| | - Lv Huang
- Rehabilitation Medicine Department, Nanchang Hongdu Hospital of Traditional Chinese Medicine, No. 264, Minde Road, Donghu District, Nanchang City, 330008, Jiangxi, China
| | - Su-Qi He
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang City, 330004, Jiangxi, China
| | - Lang-Hui Liu
- Rehabilitation Medicine Department, Nanchang Hongdu Hospital of Traditional Chinese Medicine, No. 264, Minde Road, Donghu District, Nanchang City, 330008, Jiangxi, China
| | - Song Li
- Rehabilitation Medicine Department, Nanchang Hongdu Hospital of Traditional Chinese Medicine, No. 264, Minde Road, Donghu District, Nanchang City, 330008, Jiangxi, China
| | - Zi-Chen Shao
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang City, 330004, Jiangxi, China.
| | - Jun Wang
- General Surgery Department of Trauma Center, The First Hospital of Nanchang, Nanchang City, 330008, Jiangxi, China.
| | - Ling Cheng
- Rehabilitation Medicine Department, Nanchang Hongdu Hospital of Traditional Chinese Medicine, No. 264, Minde Road, Donghu District, Nanchang City, 330008, Jiangxi, China.
| |
Collapse
|
12
|
Chen T, Karedla N, Enderlein J. Observation of E-cadherin adherens junction dynamics with metal-induced energy transfer imaging and spectroscopy. Commun Biol 2024; 7:1596. [PMID: 39613901 DOI: 10.1038/s42003-024-07281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Epithelial cadherin (E-cad) mediated cell-cell junctions play a crucial role in the establishment and maintenance of tissues and organs. In this study, we employed metal-induced energy transfer imaging and spectroscopy to investigate variations in intermembrane distance during adhesion between two model membranes adorned with E-cad. By correlating the measured intermembrane distances with the distinct E-cad junction states, we probed the dynamic behavior and diversity of E-cad junctions across different binding pathways. Our observations led to the identification of a transient intermediate state referred to as the X-dimeric state and enabled a detailed analysis of its kinetics. We discovered that the formation of the X-dimer leads to significant membrane displacement, subsequently impacting the formation of other X-dimers. These direct experimental insights into the subtle dynamics of E-cad-modified membranes and the resultant changes in intermembrane distance provide perspectives on the assembly of E-cad junctions between cells. This knowledge enhances our comprehension of tissue and organ development and may serve as a foundation for the development of innovative therapeutic strategies for diseases linked to cell-cell adhesion abnormalities.
Collapse
Affiliation(s)
- Tao Chen
- Third Institute of Physics-Biophysics, Georg August University, Göttingen, Germany.
| | - Narain Karedla
- The Rosalind Franklin Institute, Didcot, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Jörg Enderlein
- Third Institute of Physics-Biophysics, Georg August University, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), Universitätsmedizin Göttingen, Göttingen, Germany.
| |
Collapse
|
13
|
Cencer CS, Robinson KL, Tyska MJ. Loss of intermicrovillar adhesion factor CDHR2 impairs basolateral junctional complexes in transporting epithelia. Mol Biol Cell 2024; 35:br21. [PMID: 39292922 PMCID: PMC11617098 DOI: 10.1091/mbc.e24-03-0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/28/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
Transporting epithelial cells in the gut and kidney rely on protocadherin-based apical adhesion complexes to organize microvilli that extend into luminal space. In these systems, CDHR2 and CDHR5 localize to the distal ends of microvilli, where they form an intermicrovillar adhesion complex (IMAC) that links the tips of these structures, promotes the formation of a well-ordered array of protrusions, and thus maximizes apical membrane surface area. Recently, we discovered that IMACs can also form between microvilli that extend from neighboring cells, across cell-cell junctions. As an additional point of physical contact between cells, transjunctional IMACs are well positioned to impact the integrity of canonical tight and adherens junctions that form more basolaterally. To begin to test this idea, we examined cell culture and mouse models that lacked CDHR2 expression and were unable to form IMACs. CDHR2 knockout perturbed cell and junction morphology, reduced key components from tight and adherens junctions, impaired barrier function, and increased the motility of single cells within established monolayers. These results support the hypothesis that, in addition to organizing apical microvilli, IMACs provide a layer of cell-cell contact that functions in parallel with canonical tight and adherens junctions to promote epithelial functions.
Collapse
Affiliation(s)
- Caroline S. Cencer
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Kianna L. Robinson
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
14
|
Gong R, Reynolds MJ, Sun X, Alushin GM. Afadin mediates cadherin-catenin complex clustering on F-actin linked to cooperative binding and filament curvature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617332. [PMID: 39415991 PMCID: PMC11482809 DOI: 10.1101/2024.10.08.617332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The E-cadherin-β-catenin-αE-catenin (cadherin-catenin) complex couples the cytoskeletons of neighboring cells at adherens junctions (AJs) to mediate force transmission across epithelia. Mechanical force and auxiliary binding partners converge to stabilize the cadherin-catenin complex's inherently weak binding to actin filaments (F-actin) through unclear mechanisms. Here we show that afadin's coiled-coil (CC) domain and vinculin synergistically enhance the cadherin-catenin complex's F-actin engagement. The cryo-EM structure of an E-cadherin-β-catenin-αE-catenin-vinculin-afadin-CC supra-complex bound to F-actin reveals that afadin-CC bridges adjacent αE-catenin actin-binding domains along the filament, stabilizing flexible αE-catenin segments implicated in mechanical regulation. These cooperative binding contacts promote the formation of supra-complex clusters along F-actin. Additionally, cryo-EM variability analysis links supra-complex binding along individual F-actin strands to nanoscale filament curvature, a deformation mode associated with cytoskeletal forces. Collectively, this work elucidates a mechanistic framework by which vinculin and afadin tune cadherin-catenin complex-cytoskeleton coupling to support AJ function across varying mechanical regimes.
Collapse
Affiliation(s)
- Rui Gong
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Matthew J. Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Gregory M. Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
15
|
Rabino A, Awadia S, Ali N, Edson A, Garcia-Mata R. The Scribble-SGEF-Dlg1 complex regulates E-cadherin and ZO-1 stability, turnover and transcription in epithelial cells. J Cell Sci 2024; 137:jcs262181. [PMID: 39350674 PMCID: PMC11529605 DOI: 10.1242/jcs.262181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
SGEF (also known as ARHGEF26), a RhoG specific GEF, can form a ternary complex with the Scribble polarity complex proteins Scribble and Dlg1, which regulates the formation and maintenance of adherens junctions and barrier function of epithelial cells. Notably, silencing SGEF results in a dramatic downregulation of both E-cadherin and ZO-1 (also known as TJP1) protein levels. However, the molecular mechanisms involved in the regulation of this pathway are not known. Here, we describe a novel signaling pathway governed by the Scribble-SGEF-Dlg1 complex. Our results show that the three members of the ternary complex are required to maintain the stability of the apical junctions, ZO-1 protein levels and tight junction (TJ) permeability. In contrast, only SGEF is necessary to regulate E-cadherin levels. The absence of SGEF destabilizes the E-cadherin-catenin complex at the membrane, triggering a positive feedback loop that exacerbates the phenotype through the repression of E-cadherin transcription in a process that involves the internalization of E-cadherin by endocytosis, β-catenin signaling and the transcriptional repressor Slug (also known as SNAI2).
Collapse
Affiliation(s)
- Agustin Rabino
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Sahezeel Awadia
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Nabaa Ali
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Amber Edson
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|
16
|
Raab JE, Hamilton DJ, Harju TB, Huynh TN, Russo BC. Pushing boundaries: mechanisms enabling bacterial pathogens to spread between cells. Infect Immun 2024; 92:e0052423. [PMID: 38661369 PMCID: PMC11385730 DOI: 10.1128/iai.00524-23] [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: 04/26/2024] Open
Abstract
For multiple intracellular bacterial pathogens, the ability to spread directly into adjacent epithelial cells is an essential step for disease in humans. For pathogens such as Shigella, Listeria, Rickettsia, and Burkholderia, this intercellular movement frequently requires the pathogens to manipulate the host actin cytoskeleton and deform the plasma membrane into structures known as protrusions, which extend into neighboring cells. The protrusion is then typically resolved into a double-membrane vacuole (DMV) from which the pathogen quickly escapes into the cytosol, where additional rounds of intercellular spread occur. Significant progress over the last few years has begun to define the mechanisms by which intracellular bacterial pathogens spread. This review highlights the interactions of bacterial and host factors that drive mechanisms required for intercellular spread with a focus on how protrusion structures form and resolve.
Collapse
Affiliation(s)
- Julie E. Raab
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Desmond J. Hamilton
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Tucker B. Harju
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Thao N. Huynh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Brian C. Russo
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
17
|
Vagin O, Tokhtaeva E, Larauche M, Davood J, Marcus EA. Helicobacter pylori-Induced Decrease in Membrane Expression of Na,K-ATPase Leads to Gastric Injury. Biomolecules 2024; 14:772. [PMID: 39062486 PMCID: PMC11274427 DOI: 10.3390/biom14070772] [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: 05/01/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Helicobacter pylori is a highly prevalent human gastric pathogen that causes gastritis, ulcer disease, and gastric cancer. It is not yet fully understood how H. pylori injures the gastric epithelium. The Na,K-ATPase, an essential transporter found in virtually all mammalian cells, has been shown to be important for maintaining the barrier function of lung and kidney epithelia. H. pylori decreases levels of Na,K-ATPase in the plasma membrane of gastric epithelial cells, and the aim of this study was to demonstrate that this reduction led to gastric injury by impairing the epithelial barrier. Similar to H. pylori infection, the inhibition of Na,K-ATPase with ouabain decreased transepithelial electrical resistance and increased paracellular permeability in cell monolayers of human gastric cultured cells, 2D human gastric organoids, and gastric epithelium isolated from gerbils. Similar effects were caused by a partial shRNA silencing of Na,K-ATPase in human gastric organoids. Both H. pylori infection and ouabain exposure disrupted organization of adherens junctions in human gastric epithelia as demonstrated by E-cadherin immunofluorescence. Functional and structural impairment of epithelial integrity with a decrease in Na,K-ATPase amount or activity provides evidence that the H. pylori-induced downregulation of Na,K-ATPase plays a role in the complex mechanism of gastric disease induced by the bacteria.
Collapse
Affiliation(s)
- Olga Vagin
- Department of Pediatrics, DGSOM at UCLA, 10833 LeConte Ave., 12-383 MDCC, Los Angeles, CA 90095, USA; (O.V.); (E.T.)
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
| | - Elmira Tokhtaeva
- Department of Pediatrics, DGSOM at UCLA, 10833 LeConte Ave., 12-383 MDCC, Los Angeles, CA 90095, USA; (O.V.); (E.T.)
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
| | - Muriel Larauche
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, DGSOM at UCLA, 650 Charles E Young Dr. S., CHS 43-276, Los Angeles, CA 90095, USA
| | - Joshua Davood
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, DGSOM at UCLA, 650 Charles E Young Dr. S., CHS 43-276, Los Angeles, CA 90095, USA
| | - Elizabeth A. Marcus
- Department of Pediatrics, DGSOM at UCLA, 10833 LeConte Ave., 12-383 MDCC, Los Angeles, CA 90095, USA; (O.V.); (E.T.)
- VA GLAHS 11301 Wilshire Blvd, Bldg 113, Rm 324, Los Angeles, CA 90073, USA; (M.L.); (J.D.)
| |
Collapse
|
18
|
Li Q, Wang R, Xue J, Wang R, Zhang S, Kang H, Wang Y, Zhu H, Lv C. ZIF-8-Modified Black Phosphorus Nanosheets Incorporated into Injectable Dual-Component Hydrogels for Enhanced Photothermal Antibacterial and Osteogenic Activities. ACS APPLIED MATERIALS & INTERFACES 2024; 16:32058-32077. [PMID: 38872401 DOI: 10.1021/acsami.4c05298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The development of growth factor-free biomaterials for bone tissue regeneration with anti-infection and anti-inflammatory activities remains challenging. Black phosphorus nanosheets (BPNs), with distinctive attributes, including photothermal conversion and calcium ion chelation, offer potential for use in bone tissue engineering and infection prevention. However, BPNs are prone to oxidation and degradation in aqueous environments, and methods to stabilize BPNs for long-term bone repair remain insufficient. Herein, zeolitic imidazolate framework-8 (ZIF-8) was used to stabilize BPNs via in situ crystallization onto the surface of BPNs (BP@ZIF-8 nanocomposite). A novel injectable dual-component hydrogel comprising gelatin methacryloyl (GelMA) and methacrylate-modified hyaluronic acid (HAMA) was used as a BP@ZIF-8 nanocomposite carrier (GelMA/HAMA/BP@ZIF-8). The BP@ZIF-8 nanocomposite could effectively protect internal BPNs from oxidation and enhance the long-term photothermal performance of the hydrogel in both in vitro and in vivo settings. The GelMA/HAMA/BP@ZIF-8 hydrogel was injectable and exhibited outstanding performance for photothermal conversion, mechanical strength, and biodegradability, as well as excellent photothermal antibacterial activity against Staphylococcus aureus and Escherichia coli in vitro and in an in vivo rat model. The GelMA/HAMA/BP@ZIF-8 hydrogel also provided a microenvironment conducive to osteogenic differentiation, promoting the transformation of M2 macrophages and inhibiting inflammatory responses. Furthermore, the hydrogel promoted bone regeneration and had a synergistic effect with near-infrared irradiation in a rat skull-defect model. Transcriptome sequencing analysis revealed that the PI3K-AKT- and calcium-signaling pathways may be involved in promoting osteogenic differentiation induced by the GH-BZ hydrogel. This study presents an innovative, multifaceted solution to the challenges of bone tissue regeneration with antibacterial and anti-inflammatory effects, providing insights into the design of smart biomaterials with dual therapeutic capabilities.
Collapse
Affiliation(s)
- Quan Li
- Emergency Department, The State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou 571199, China
| | - Ruijie Wang
- Emergency Department, The State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Jinfang Xue
- Emergency Department, The State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Ruiyu Wang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Shun Zhang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Hai Kang
- Emergency Department, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Yang Wang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Huadong Zhu
- Emergency Department, The State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Chuanzhu Lv
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou 571199, China
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
19
|
Xie B, Wu T, Hong D, Lu Z. Comprehensive landscape of junctional genes and their association with overall survival of patients with lung adenocarcinoma. Front Mol Biosci 2024; 11:1380384. [PMID: 38841188 PMCID: PMC11150628 DOI: 10.3389/fmolb.2024.1380384] [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: 02/01/2024] [Accepted: 04/22/2024] [Indexed: 06/07/2024] Open
Abstract
Objectives Junctional proteins are involved in tumorigenesis. Therefore, this study aimed to investigate the association between junctional genes and the prognosis of patients with lung adenocarcinoma (LUAD). Methods Transcriptome, mutation, and clinical data were retrieved from The Cancer Genome Atlas (TCGA). "Limma" was used to screen differentially expressed genes. Moreover, Kaplan-Meier survival analysis was used to identify junctional genes associated with LUAD prognosis. The junctional gene-related risk score (JGRS) was generated based on multivariate Cox regression analysis. An overall survival (OS) prediction model combining the JGRS and clinicopathological properties was proposed using a nomogram and further validated in the Gene Expression Omnibus (GEO) LUAD cohort. Results To our knowledge, this study is the first to demonstrate the correlation between the mRNA levels of 14 junctional genes (CDH15, CDH17, CDH24, CLDN6, CLDN12, CLDN18, CTNND2, DSG2, ITGA2, ITGA8, ITGA11, ITGAL, ITGB4, and PKP3) and clinical outcomes of patients with LUAD. The JGRS was generated based on these 14 genes, and a higher JGRS was associated with older age, higher stage levels, and lower immune scores. Thus, a prognostic prediction nomogram was proposed based on the JGRS. Internal and external validation showed the good performance of the prediction model. Mechanistically, JGRS was associated with cell proliferation and immune regulatory pathways. Mutational analysis revealed that more somatic mutations occurred in the high-JGRS group than in the low-JGRS group. Conclusion The association between junctional genes and OS in patients with LUAD demonstrated by our "TCGA filtrating and GEO validating" model revealed a new function of junctional genes.
Collapse
Affiliation(s)
- Bin Xie
- School of Information Science and Technology, Hangzhou Normal University, Hangzhou, China
| | - Ting Wu
- School of Information Science and Technology, Hangzhou Normal University, Hangzhou, China
| | - Duiguo Hong
- Jincheng Community Health Service Center, Hangzhou, China
| | - Zhe Lu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
- School of Basic Medicine, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
20
|
Ilnitskaya AS, Litovka NI, Rubtsova SN, Zhitnyak IY, Gloushankova NA. Actin Cytoskeleton Remodeling Accompanied by Redistribution of Adhesion Proteins Drives Migration of Cells in Different EMT States. Cells 2024; 13:780. [PMID: 38727316 PMCID: PMC11083118 DOI: 10.3390/cells13090780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process during which epithelial cells lose epithelial characteristics and gain mesenchymal features. Here, we used several cell models to study migratory activity and redistribution of cell-cell adhesion proteins in cells in different EMT states: EGF-induced EMT of epithelial IAR-20 cells; IAR-6-1 cells with a hybrid epithelial-mesenchymal phenotype; and their more mesenchymal derivatives, IAR-6-1-DNE cells lacking adherens junctions. In migrating cells, the cell-cell adhesion protein α-catenin accumulated at the leading edges along with ArpC2/p34 and α-actinin. Suppression of α-catenin shifted cell morphology from fibroblast-like to discoid and attenuated cell migration. Expression of exogenous α-catenin in MDA-MB-468 cells devoid of α-catenin drastically increased their migratory capabilities. The Y654 phosphorylated form of β-catenin was detected at integrin adhesion complexes (IACs). Co-immunoprecipitation studies indicated that α-catenin and pY654-β-catenin were associated with IAC proteins: vinculin, zyxin, and α-actinin. Taken together, these data suggest that in cells undergoing EMT, catenins not participating in assembly of adherens junctions may affect cell migration.
Collapse
Affiliation(s)
- Alla S. Ilnitskaya
- Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, 115478 Moscow, Russia; (A.S.I.); (N.I.L.); (S.N.R.); (I.Y.Z.)
| | - Nikita I. Litovka
- Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, 115478 Moscow, Russia; (A.S.I.); (N.I.L.); (S.N.R.); (I.Y.Z.)
| | - Svetlana N. Rubtsova
- Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, 115478 Moscow, Russia; (A.S.I.); (N.I.L.); (S.N.R.); (I.Y.Z.)
| | - Irina Y. Zhitnyak
- Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, 115478 Moscow, Russia; (A.S.I.); (N.I.L.); (S.N.R.); (I.Y.Z.)
- Department of Molecular Genetics, University of Toronto, 661 University Ave, MaRS West, Toronto, ON 5MG 1M1, Canada
| | - Natalya A. Gloushankova
- Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, 115478 Moscow, Russia; (A.S.I.); (N.I.L.); (S.N.R.); (I.Y.Z.)
| |
Collapse
|
21
|
Luo C, Xu Y, Zhang J, Tian Q, Guo Y, Li N, Feng Y, Xu R, Xiao L. Cryptosporidium parvum disrupts intestinal epithelial barrier in neonatal mice through downregulation of cell junction molecules. PLoS Negl Trop Dis 2024; 18:e0012212. [PMID: 38787872 PMCID: PMC11156435 DOI: 10.1371/journal.pntd.0012212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/06/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Cryptosporidium spp. cause watery diarrhea in humans and animals, especially in infants and neonates. They parasitize the apical surface of the epithelial cells in the intestinal lumen. However, the pathogenesis of Cryptosporidium-induced diarrhea is not fully understood yet. METHODOLOGY/PRINCIPAL FINDINGS In this study, we infected C57BL/6j neonatal mice with C. parvum IIa and IId subtypes, and examined oocyst burden, pathological changes, and intestinal epithelial permeability during the infection. In addition, transcriptomic analyses were used to study the mechanism of diarrhea induced by the C. parvum IId subtype. The neonatal mice were sensitive to both C. parvum IIa and IId infection, but the IId subtype caused a wide oocyst shedding window and maintained the high oocyst burden in the mice compared with the IIa subtype. In addition, the mice infected with C. parvum IId resulted in severe intestinal damage at the peak of infection, leading to increased permeability of the epithelial barrier. The KEGG, GO and GSEA analyses revealed that the downregulation of adherens junction and cell junction molecules at 11 dpi. Meanwhile, E-cadherin, which is associated with adherens junction, was reduced at the protein level in mouse ileum at peak and late infection. CONCLUSIONS/SIGNIFICANCE C. parvum IId infection causes more severe pathological damage than C. parvum IIa infection in neonatal mice. Furthermore, the impairment of the epithelial barrier during C. parvum IId infection results from the downregulation of intestinal junction proteins.
Collapse
Affiliation(s)
- Chaowei Luo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yanhua Xu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Jie Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Qing Tian
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yaqiong Guo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Na Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yaoyu Feng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Rui Xu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Lihua Xiao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
22
|
Rabino A, Awadia S, Ali N, Edson A, Garcia-Mata R. The Scribble/SGEF/Dlg1 complex regulates the stability of apical junctions in epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586884. [PMID: 38585765 PMCID: PMC10996629 DOI: 10.1101/2024.03.26.586884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
SGEF, a RhoG specific GEF, can form a ternary complex with the Scribble polarity complex proteins Scribble and Dlg1, which regulates the formation and maintenance of adherens junctions and barrier function of epithelial cells. Notably, silencing SGEF results in a dramatic downregulation of the expression of both E-cadherin and ZO-1. However, the molecular mechanisms involved in the regulation of this pathway are not known. Here, we describe a novel signaling pathway governed by the Scribble/SGEF/Dlg1 complex. Our results show that an intact ternary complex is required to maintain the stability of the apical junctions, the expression of ZO-1, and TJ permeability. In contrast, only SGEF is necessary to regulate E-cadherin expression. The absence of SGEF destabilizes the E-cadherin/catenin complex at the membrane, triggering a positive feedback loop that exacerbates the phenotype through the repression of E-cadherin transcription in a process that involves the internalization of E-cadherin by endocytosis, β-catenin signaling and the transcriptional repressor Slug.
Collapse
Affiliation(s)
- Agustin Rabino
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Sahezeel Awadia
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Nabaa Ali
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Amber Edson
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
23
|
Cencer CS, Robinson KL, Tyska MJ. Loss of intermicrovillar adhesion impairs basolateral junctional complexes in transporting epithelia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585733. [PMID: 38562895 PMCID: PMC10983982 DOI: 10.1101/2024.03.19.585733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Transporting epithelial cells in the gut and kidney rely on protocadherin-based apical adhesion complexes to organize microvilli that extend into the luminal space. In these systems, CDHR2 and CDHR5 localize to the distal ends of microvilli, where they form an intermicrovillar adhesion complex (IMAC) that links the tips of these structures, promotes the formation of a well-ordered array of protrusions, and in turn maximizes apical membrane surface area. Recently, we discovered that IMACs can also form between microvilli that extend from neighboring cells, across cell-cell junctions. As an additional point of physical contact between cells, transjunctional IMACs are well positioned to impact the integrity of canonical tight and adherens junctions that form more basolaterally. Here, we sought to test this idea using cell culture and mouse models that lacked CDHR2 expression and were unable to form IMACs. CDHR2 knockout perturbed cell and junction morphology, led to loss of key components from tight and adherens junctions, and impaired barrier function and wound healing. These results indicate that, in addition to organizing apical microvilli, IMACs provide a layer of cell-cell contact that functions in parallel with canonical tight and adherens junctions to support the physiological functions of transporting epithelia.
Collapse
|
24
|
Troyanovsky RB, Indra I, Troyanovsky SM. Characterization of early and late events of adherens junction assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583373. [PMID: 38496678 PMCID: PMC10942379 DOI: 10.1101/2024.03.04.583373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Cadherins are transmembrane adhesion receptors. Cadherin ectodomains form adhesive 2D clusters through cooperative trans and cis interactions, whereas its intracellular region interacts with specific cytosolic proteins, termed catenins, to anchor the cadherin-catenin complex (CCC) to the actin cytoskeleton. How these two types of interactions are coordinated in the formation of specialized cell-cell adhesions, adherens junctions (AJ), remains unclear. We focus here on the role of the actin-binding domain of α-catenin (αABD) by showing that the interaction of αABD with actin generates actin-bound CCC oligomers (CCC/actin strands) incorporating up to six CCCs. The strands are primarily formed on the actin-rich cell protrusions. Once in cell-cell interface, the strands become involved in cadherin ectodomain clustering. Such combination of the extracellular and intracellular oligomerizations gives rise to the composite oligomers, trans CCC/actin clusters. To mature, these clusters then rearrange their actin filaments using several redundant pathways, two of which are characterized here: one depends on the α-catenin-associated protein, vinculin and the second one depends on the unstructured C-terminus of αABD. Thus, AJ assembly proceeds through spontaneous formation of trans CCC/actin clusters and their successive reorganization.
Collapse
Affiliation(s)
- Regina B Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
| | - Indrajyoti Indra
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
| | - Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
- Department of Cell & Developmental Biology, The Feinberg School of Medicine, Chicago, IL 60614
| |
Collapse
|
25
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
26
|
Tsai YX, Chang NE, Reuter K, Chang HT, Yang TJ, von Bülow S, Sehrawat V, Zerrouki N, Tuffery M, Gecht M, Grothaus IL, Colombi Ciacchi L, Wang YS, Hsu MF, Khoo KH, Hummer G, Hsu STD, Hanus C, Sikora M. Rapid simulation of glycoprotein structures by grafting and steric exclusion of glycan conformer libraries. Cell 2024; 187:1296-1311.e26. [PMID: 38428397 DOI: 10.1016/j.cell.2024.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 10/18/2023] [Accepted: 01/22/2024] [Indexed: 03/03/2024]
Abstract
Most membrane proteins are modified by covalent addition of complex sugars through N- and O-glycosylation. Unlike proteins, glycans do not typically adopt specific secondary structures and remain very mobile, shielding potentially large fractions of protein surface. High glycan conformational freedom hinders complete structural elucidation of glycoproteins. Computer simulations may be used to model glycosylated proteins but require hundreds of thousands of computing hours on supercomputers, thus limiting routine use. Here, we describe GlycoSHIELD, a reductionist method that can be implemented on personal computers to graft realistic ensembles of glycan conformers onto static protein structures in minutes. Using molecular dynamics simulation, small-angle X-ray scattering, cryoelectron microscopy, and mass spectrometry, we show that this open-access toolkit provides enhanced models of glycoprotein structures. Focusing on N-cadherin, human coronavirus spike proteins, and gamma-aminobutyric acid receptors, we show that GlycoSHIELD can shed light on the impact of glycans on the conformation and activity of complex glycoproteins.
Collapse
Affiliation(s)
- Yu-Xi Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Ning-En Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Klaus Reuter
- Max Planck Computing and Data Facility, 85748 Garching, Germany
| | - Hao-Ting Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Tzu-Jing Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Sören von Bülow
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany
| | - Vidhi Sehrawat
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany; Malopolska Centre of Biotechnology, Jagiellonian University, 31-007 Kraków, Poland
| | - Noémie Zerrouki
- Institute of Psychiatry and Neurosciences of Paris, Inserm UMR1266, Université Paris-Cité, 75014 Paris, France
| | - Matthieu Tuffery
- Institute of Psychiatry and Neurosciences of Paris, Inserm UMR1266, Université Paris-Cité, 75014 Paris, France
| | - Michael Gecht
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany
| | - Isabell Louise Grothaus
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, Bremen Center for Computational Materials Science and MAPEX Center for Materials and Processes, University of Bremen, 28359 Bremen, Germany
| | - Lucio Colombi Ciacchi
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, Bremen Center for Computational Materials Science and MAPEX Center for Materials and Processes, University of Bremen, 28359 Bremen, Germany
| | - Yong-Sheng Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Min-Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany; Institute of Biophysics, Goethe University, 60438 Frankfurt, Germany
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan; International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM(2)), Hiroshima University, Hiroshima 739-8526, Japan.
| | - Cyril Hanus
- Institute of Psychiatry and Neurosciences of Paris, Inserm UMR1266, Université Paris-Cité, 75014 Paris, France; GHU Psychiatrie et Neurosciences de Paris, 75014 Paris, France.
| | - Mateusz Sikora
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany; Malopolska Centre of Biotechnology, Jagiellonian University, 31-007 Kraków, Poland.
| |
Collapse
|
27
|
Mira-Osuna M, Borgne RL. Assembly, dynamics and remodeling of epithelial cell junctions throughout development. Development 2024; 151:dev201086. [PMID: 38205947 DOI: 10.1242/dev.201086] [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: 01/12/2024]
Abstract
Cell junctions play key roles in epithelial integrity. During development, when epithelia undergo extensive morphogenesis, these junctions must be remodeled in order to maintain mechanochemical barriers and ensure the cohesion of the tissue. In this Review, we present a comprehensive and integrated description of junctional remodeling mechanisms in epithelial cells during development, from embryonic to adult epithelia. We largely focus on Drosophila, as quantitative analyses in this organism have provided a detailed characterization of the molecular mechanisms governing cell topologies, and discuss the conservation of these mechanisms across metazoans. We consider how changes at the molecular level translate to tissue-scale irreversible deformations, exploring the composition and assembly of cellular interfaces to unveil how junctions are remodeled to preserve tissue homeostasis during cell division, intercalation, invagination, ingression and extrusion.
Collapse
Affiliation(s)
- Marta Mira-Osuna
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS UMR 6290, F-35000 Rennes, France
| | - Roland Le Borgne
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS UMR 6290, F-35000 Rennes, France
| |
Collapse
|
28
|
Koff M, Monagas-Valentin P, Novikov B, Chandel I, Panin V. Protein O-mannosylation: one sugar, several pathways, many functions. Glycobiology 2023; 33:911-926. [PMID: 37565810 PMCID: PMC10859634 DOI: 10.1093/glycob/cwad067] [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: 07/16/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023] Open
Abstract
Recent research has unveiled numerous important functions of protein glycosylation in development, homeostasis, and diseases. A type of glycosylation taking the center stage is protein O-mannosylation, a posttranslational modification conserved in a wide range of organisms, from yeast to humans. In animals, protein O-mannosylation plays a crucial role in the nervous system, whereas protein O-mannosylation defects cause severe neurological abnormalities and congenital muscular dystrophies. However, the molecular and cellular mechanisms underlying protein O-mannosylation functions and biosynthesis remain not well understood. This review outlines recent studies on protein O-mannosylation while focusing on the functions in the nervous system, summarizes the current knowledge about protein O-mannosylation biosynthesis, and discusses the pathologies associated with protein O-mannosylation defects. The evolutionary perspective revealed by studies in the Drosophila model system are also highlighted. Finally, the review touches upon important knowledge gaps in the field and discusses critical questions for future research on the molecular and cellular mechanisms associated with protein O-mannosylation functions.
Collapse
Affiliation(s)
- Melissa Koff
- Department of Biochemistry and Biophysics, AgriLife Research, Texas A&M University, College Station, College Station, TX 77843, United States
| | - Pedro Monagas-Valentin
- Department of Biochemistry and Biophysics, AgriLife Research, Texas A&M University, College Station, College Station, TX 77843, United States
| | - Boris Novikov
- Department of Biochemistry and Biophysics, AgriLife Research, Texas A&M University, College Station, College Station, TX 77843, United States
| | - Ishita Chandel
- Department of Biochemistry and Biophysics, AgriLife Research, Texas A&M University, College Station, College Station, TX 77843, United States
| | - Vladislav Panin
- Department of Biochemistry and Biophysics, AgriLife Research, Texas A&M University, College Station, College Station, TX 77843, United States
| |
Collapse
|
29
|
Mukherjee S, Goswami S, Dash S, Samanta D. Structural basis of molecular recognition among classical cadherins mediating cell adhesion. Biochem Soc Trans 2023; 51:2103-2115. [PMID: 37970977 DOI: 10.1042/bst20230356] [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: 09/01/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Cadherins are type-I membrane glycoproteins that primarily participate in calcium-dependent cell adhesion and homotypic cell sorting in various stages of embryonic development. Besides their crucial role in cellular and physiological processes, increasing studies highlight their involvement in pathophysiological functions ranging from cancer progression and metastasis to being entry receptors for pathogens. Cadherins mediate these cellular processes through homophilic, as well as heterophilic interactions (within and outside the superfamily) by their membrane distal ectodomains. This review provides an in-depth structural perspective of molecular recognition among type-I and type-II classical cadherins. Furthermore, this review offers structural insights into different dimeric assemblies like the 'strand-swap dimer' and 'X-dimer' as well as mechanisms relating these dimer forms like 'two-step adhesion' and 'encounter complex'. Alongside providing structural details, this review connects structural studies to bond mechanics merging crystallographic and single-molecule force spectroscopic findings. Finally, the review discusses the recent discoveries on dimeric intermediates that uncover prospects of further research beyond two-step adhesion.
Collapse
Affiliation(s)
- Sarbartha Mukherjee
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Saumyadeep Goswami
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Sagarika Dash
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Dibyendu Samanta
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| |
Collapse
|
30
|
Schmidt A, Finegan T, Häring M, Kong D, Fletcher AG, Alam Z, Grosshans J, Wolf F, Peifer M. Polychaetoid/ZO-1 strengthens cell junctions under tension while localizing differently than core adherens junction proteins. Mol Biol Cell 2023; 34:ar81. [PMID: 37163320 PMCID: PMC10398881 DOI: 10.1091/mbc.e23-03-0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023] Open
Abstract
During embryonic development, dramatic cell shape changes and movements reshape the embryonic body plan. These require robust but dynamic linkage between the cell-cell adherens junctions and the force-generating actomyosin cytoskeleton. Our view of this linkage has evolved, and we now realize linkage is mediated by mechanosensitive multiprotein complexes assembled via multivalent connections. Here we combine genetic, cell biological, and modeling approaches to define the mechanism of action and functions of an important player, Drosophila polychaetoid, homologue of mammalian ZO-1. Our data reveal that Pyd reinforces cell junctions under elevated tension, and facilitates cell rearrangements. Pyd is important to maintain junctional contractility and in its absence cell rearrangements stall. We next use structured illumination microscopy to define the molecular architecture of cell-cell junctions during these events. The cadherin-catenin complex and Cno both localize to puncta along the junctional membrane, but are differentially enriched in different puncta. Pyd, in contrast, exhibits a distinct localization to strands that extend out from the region occupied by core junction proteins. We then discuss the implications for the protein network at the junction-cytoskeletal interface, suggesting different proteins localize and function in distinct ways, perhaps in distinct subcomplexes, but combine to produce robust connections.
Collapse
Affiliation(s)
- Anja Schmidt
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | - Tara Finegan
- Department of Biology, University of Rochester, Rochester, New York 14627-0211
| | - Matthias Häring
- Göttingen Campus Institute for Dynamics of Biological Networks, Georg August University, 37075 Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
- Institute for Dynamics of Complex Systems, Georg August University, 37077 Göttingen, Germany
| | - Deqing Kong
- Department of Biology, Philipps University, 35043 Marburg, Germany
| | - Alexander G Fletcher
- School of Mathematics and Statistics and Bateson Centre, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Zuhayr Alam
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | - Jörg Grosshans
- Department of Biology, Philipps University, 35043 Marburg, Germany
| | - Fred Wolf
- Göttingen Campus Institute for Dynamics of Biological Networks, Georg August University, 37075 Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
- Institute for Dynamics of Complex Systems, Georg August University, 37077 Göttingen, Germany
| | - Mark Peifer
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| |
Collapse
|
31
|
Lin WH, Cooper LM, Anastasiadis PZ. Cadherins and catenins in cancer: connecting cancer pathways and tumor microenvironment. Front Cell Dev Biol 2023; 11:1137013. [PMID: 37255594 PMCID: PMC10225604 DOI: 10.3389/fcell.2023.1137013] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023] Open
Abstract
Cadherin-catenin complexes are integral components of the adherens junctions crucial for cell-cell adhesion and tissue homeostasis. Dysregulation of these complexes is linked to cancer development via alteration of cell-autonomous oncogenic signaling pathways and extrinsic tumor microenvironment. Advances in multiomics have uncovered key signaling events in multiple cancer types, creating a need for a better understanding of the crosstalk between cadherin-catenin complexes and oncogenic pathways. In this review, we focus on the biological functions of classical cadherins and associated catenins, describe how their dysregulation influences major cancer pathways, and discuss feedback regulation mechanisms between cadherin complexes and cellular signaling. We discuss evidence of cross regulation in the following contexts: Hippo-Yap/Taz and receptor tyrosine kinase signaling, key pathways involved in cell proliferation and growth; Wnt, Notch, and hedgehog signaling, key developmental pathways involved in human cancer; as well as TGFβ and the epithelial-to-mesenchymal transition program, an important process for cancer cell plasticity. Moreover, we briefly explore the role of cadherins and catenins in mechanotransduction and the immune tumor microenvironment.
Collapse
|
32
|
Schmidt A, Finegan T, Häring M, Kong D, Fletcher AG, Alam Z, Grosshans J, Wolf F, Peifer M. Polychaetoid/ZO-1 strengthens cell junctions under tension while localizing differently than core adherens junction proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530634. [PMID: 36909597 PMCID: PMC10002719 DOI: 10.1101/2023.03.01.530634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
During embryonic development dramatic cell shape changes and movements re-shape the embryonic body plan. These require robust but dynamic linkage between the cell-cell adherens junctions and the force-generating actomyosin cytoskeleton. Our view of this linkage has evolved, and we now realize linkage is mediated by a mechanosensitive multiprotein complex assembled via multivalent connections. Here we combine genetic, cell biological and modeling approaches to define the mechanism of action and functions of an important player, Drosophila Polychaetoid, homolog of mammalian ZO-1. Our data reveal that Pyd reinforces cell junctions under elevated tension, and facilitates cell rearrangements. Pyd is important to maintain junctional contractility and in its absence cell rearrangements stall. We next use structured illumination microscopy to define the molecular architecture of cell-cell junctions during these events. The cadherin-catenin complex and Cno both localize to puncta along the junctional membrane, but are differentially enriched in different puncta. Pyd, in contrast, exhibits a distinct localization to strands that extend out from the region occupied by core junction proteins. We then discuss the implications for the protein network at the junction-cytoskeletal interface, suggesting different proteins localize and function in distinct ways but combine to produce robust connections.
Collapse
Affiliation(s)
- Anja Schmidt
- Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA
| | - Tara Finegan
- Department of Biology, University of Rochester, Rochester, New York, USA 14627-0211
| | - Matthias Häring
- Göttingen Campus Institute for Dynamics of Biological Networks, Georg August University, Hermann Rein Str. 3, 37075 Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, Am Faßberg 17, 37077 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Hermann Rein St. 3, 37075 Göttingen, German
- Institute for Dynamics of Complex Systems, Georg August University, Friedrich Hund Pl. 1, 37077 Göttingen, Germany
| | - Deqing Kong
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Alexander G Fletcher
- School of Mathematics and Statistics & Bateson Centre, University of Sheffield, Sheffield, UK
| | - Zuhayr Alam
- Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA
| | - Jörg Grosshans
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Fred Wolf
- Göttingen Campus Institute for Dynamics of Biological Networks, Georg August University, Hermann Rein Str. 3, 37075 Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, Am Faßberg 17, 37077 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Hermann Rein St. 3, 37075 Göttingen, German
- Institute for Dynamics of Complex Systems, Georg August University, Friedrich Hund Pl. 1, 37077 Göttingen, Germany
| | - Mark Peifer
- Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA
| |
Collapse
|
33
|
Munoz C, Jayanthi S, Ladenheim B, Cadet JL. Compulsive methamphetamine self-administration in the presence of adverse consequences is associated with increased hippocampal mRNA expression of cellular adhesion molecules. Front Mol Neurosci 2023; 15:1104657. [PMID: 36710935 PMCID: PMC9880890 DOI: 10.3389/fnmol.2022.1104657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/19/2022] [Indexed: 01/15/2023] Open
Abstract
Methamphetamine (METH) is a popular but harmful psychostimulant. METH use disorder (MUD) is characterized by compulsive and continued use despite adverse life consequences. METH users experience impairments in learning and memory functions that are thought to be secondary to METH-induced abnormalities in the hippocampus. Recent studies have reported that about 50% of METH users develop MUD, suggesting that there may be differential molecular effects of METH between the brains of individuals who met criteria for addiction and those who did not after being exposed to the drug. The present study aimed at identifying potential transcriptional differences between compulsive and non-compulsive METH self-administering male rats by measuring global gene expression changes in the hippocampus using RNA sequencing. Herein, we used a model of METH self-administration (SA) accompanied by contingent foot-shock punishment. This approach led to the separation of animals into shock-resistant rats (compulsive) that continued to take METH and shock-sensitive rats (non-compulsive) that suppressed their METH intake in the presence of punished METH taking. Rats were euthanized 2 h after the last METH SA plus foot-shock session. Their hippocampi were immediately removed, frozen, and used later for RNA sequencing and qRT-PCR analyses. RNA sequencing analyses revealed differential expression of mRNAs encoding cell adhesion molecules (CAMs) between the two rat phenotypes. qRT-PCR analyses showed significant higher levels of Cdh1, Glycam1, and Mpzl2 mRNAs in the compulsive rats in comparison to non-compulsive rats. The present results implicate altered CAM expression in the hippocampus in the behavioral manifestations of continuous compulsive METH taking in the presence of adverse consequences. Our results raise the novel possibility that altered CAM expression might play a role in compulsive METH taking and the cognitive impairments observed in MUD patients.
Collapse
|