1
|
Rätze MAK, Enserink LNFL, Ishiyama N, van Kempen S, Veltman CHJ, Nijman IJ, Haakma WE, Caldas C, Bernards R, van Diest PJ, Christgen M, Koorman T, Derksen PWB. Afadin loss induces breast cancer metastasis through destabilisation of E-cadherin to F-actin linkage. J Pathol 2025; 266:26-39. [PMID: 40026293 PMCID: PMC11985701 DOI: 10.1002/path.6394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/18/2024] [Accepted: 12/16/2024] [Indexed: 03/05/2025]
Abstract
Afadin is a multimodal scaffolding protein with essential functions in cell-cell adhesion. Although its loss of expression has been linked to breast cancer invasion and metastasis, the underlying mechanisms driving tumour progression upon mutational Afadin (AFDN) loss in breast cancers remains unclear. In the current study we identified a somatic frameshift AFDN mutation (p.Lys630fs) in an invasive breast cancer sample that coincides with loss of Afadin protein expression. Functional studies in E-cadherin-expressing breast cancer cells show that Afadin loss leads to immature and aberrant adherens junction (AJ) formation. The lack of AJ maturation results in a noncohesive cellular phenotype accompanied by Actomyosin-dependent anoikis resistance, which are classical progression hallmarks of single-cell breast cancer invasion. Reconstitution experiments using Afadin truncates show that proper F-actin organisation and epithelial cell-cell adhesion critically depend on the Coiled-Coil domain of Afadin but not on the designated C-terminal F-actin binding domain. Mouse xenograft experiments based on cell lines and primary patient-derived breast cancer organoids demonstrate that Afadin loss induces single-cell lobular-type invasion phenotypes and overt dissemination to the lungs and the peritoneum. In short, Afadin is a metastasis suppressor for breast cancer through stabilisation and maturation of a mechanical E-cadherin to F-actin outside-in link. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Max AK Rätze
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Lotte NFL Enserink
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Sven van Kempen
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Isaac J Nijman
- Center for Molecular Medicine, Cancer Genomics Netherlands, Department of GeneticsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Wisse E Haakma
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Department of OncologyUniversity of CambridgeCambridgeUK
| | - René Bernards
- Division of Molecular Carcinogenesis, Center for Biomedical Genetics and Cancer Genomics CentreThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Paul J van Diest
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Thijs Koorman
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Patrick WB Derksen
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
2
|
Yadav SS, Srinivasan K, Sharma SS, Datusalia AK. Decoding the Nectin Interactome: Implications for Brain Development, Plasticity, and Neurological Disorders. ACS Chem Neurosci 2025; 16:1000-1020. [PMID: 40025835 DOI: 10.1021/acschemneuro.5c00069] [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] [Indexed: 03/04/2025] Open
Abstract
The nectin family of cell adhesion molecules (CAMs) comprising nectins and nectin-like molecules has emerged as a key regulator of various pivotal neural processes, including neuronal development, migration, synapse formation, and plasticity. Nectins engage in homophilic and heterophilic interactions to mediate cell-cell adhesion, contributing to the establishment and maintenance of neural circuits. Their extracellular domains facilitate trans-synaptic interactions, while intracellular domains participate in signaling cascades influencing cytoskeletal dynamics and synaptic function. The exhibition of distinct localization patterns in neurons, astrocytes, and the blood-brain barrier underscores their diverse roles in the brain. The dysregulation of nectins has been implicated in several neurological disorders, such as neurodevelopmental disorders, depression, schizophrenia, and Alzheimer's disease. This review examines the structural and functional characteristics of nectins and their distribution and molecular mechanisms governing neural connectivity and cognition. It further discusses experimental studies unraveling nectin-mediated pathophysiology and potential therapeutic interventions targeting nectin-related pathways. Collectively, this comprehensive analysis highlights the significance of nectins in brain development, function, and disorders, paving the way for future research directions and clinical implications.
Collapse
Affiliation(s)
- Shreyash Santosh Yadav
- Molecular NeuroTherapeutics Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh 226002, India
| | - Krishnamoorthy Srinivasan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Ashok Kumar Datusalia
- Molecular NeuroTherapeutics Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Uttar Pradesh 226002, India
| |
Collapse
|
3
|
Sakakibara S, Sakane A, Sasaki T, Shinohara M, Maruo T, Miyata M, Mizutani K, Takai Y. Identification of lysophosphatidic acid in serum as a factor that promotes epithelial apical junctional complex organization. J Biol Chem 2022; 298:102426. [PMID: 36030821 PMCID: PMC9520027 DOI: 10.1016/j.jbc.2022.102426] [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: 04/26/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
The apical junctional complex (AJC) consists of adherens junctions (AJs) and tight junctions and regulates epithelial integrity and remodeling. However, it is unclear how AJC organization is regulated based on environmental cues. We found here using cultured EpH4 mouse mammary epithelial cells that fetal bovine serum (FBS) in a culture medium showed an activity to promote AJC organization and that FBS showed an activity to promote tight junction formation even in the absence of AJ proteins, such as E-cadherin, αE-catenin, and afadin. Furthermore, we purified the individual factor responsible for these functions from FBS and identified this molecule as lysophosphatidic acid (LPA). In validation experiments, purified LPA elicited the same activity as FBS. In addition, we found that the AJC organization–promoting activity of LPA was mediated through the LPA receptor 1/5 via diacylglycerol–novel PKC and Rho–ROCK pathway activation in a mutually independent, but complementary, manner. We demonstrated that the Rho–ROCK pathway activation–mediated AJC organization was independent of myosin II-induced actomyosin contraction, although this signaling pathway was previously shown to induce myosin II activation. These findings are in contrast to the literature, as previous results suggested an AJC organization–disrupting activity of LPA. The present results indicate that LPA in serum has an AJC organization–promoting activity in a manner dependent on or independent of AJ proteins.
Collapse
Affiliation(s)
- Shotaro Sakakibara
- Department of Biochemistry, Tokushima University Graduate School of Medicine, Tokushima 770-8503, Japan; Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
| | - Ayuko Sakane
- Department of Biochemistry, Tokushima University Graduate School of Medicine, Tokushima 770-8503, Japan; Department of Interdisciplinary Researches for Medicine and Photonics, Institute of Post-LED Photonics, Tokushima University, Tokushima 770-8503, Japan.
| | - Takuya Sasaki
- Department of Biochemistry, Tokushima University Graduate School of Medicine, Tokushima 770-8503, Japan
| | - Masakazu Shinohara
- Division of Epidemiology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan; The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Tomohiko Maruo
- Department of Biochemistry, Tokushima University Graduate School of Medicine, Tokushima 770-8503, Japan; Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
| | - Muneaki Miyata
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
| | - Kiyohito Mizutani
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan.
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan.
| |
Collapse
|
4
|
Wang J, Chen X. Junctional Adhesion Molecules: Potential Proteins in Atherosclerosis. Front Cardiovasc Med 2022; 9:888818. [PMID: 35872908 PMCID: PMC9302484 DOI: 10.3389/fcvm.2022.888818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Junctional adhesion molecules (JAMs) are cell-cell adhesion molecules of the immunoglobulin superfamily and are involved in the regulation of diverse atherosclerosis-related processes such as endothelial barrier maintenance, leucocytes transendothelial migration, and angiogenesis. To combine and further broaden related results, this review concluded the recent progress in the roles of JAMs and predicted future studies of JAMs in the development of atherosclerosis.
Collapse
Affiliation(s)
- Junqi Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaoping Chen,
| |
Collapse
|
5
|
Vasquez CG, de la Serna EL, Dunn AR. How cells tell up from down and stick together to construct multicellular tissues - interplay between apicobasal polarity and cell-cell adhesion. J Cell Sci 2021; 134:272658. [PMID: 34714332 DOI: 10.1242/jcs.248757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Polarized epithelia define a topological inside and outside, and hence constitute a key evolutionary innovation that enabled the construction of complex multicellular animal life. Over time, this basic function has been elaborated upon to yield the complex architectures of many of the organs that make up the human body. The two processes necessary to yield a polarized epithelium, namely regulated adhesion between cells and the definition of the apicobasal (top-bottom) axis, have likewise undergone extensive evolutionary elaboration, resulting in multiple sophisticated protein complexes that contribute to both functions. Understanding how these components function in combination to yield the basic architecture of a polarized cell-cell junction remains a major challenge. In this Review, we introduce the main components of apicobasal polarity and cell-cell adhesion complexes, and outline what is known about their regulation and assembly in epithelia. In addition, we highlight studies that investigate the interdependence between these two networks. We conclude with an overview of strategies to address the largest and arguably most fundamental unresolved question in the field, namely how a polarized junction arises as the sum of its molecular parts.
Collapse
Affiliation(s)
- Claudia G Vasquez
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Eva L de la Serna
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA.,Biophysics Program, Stanford University, Stanford, CA 94305, USA.,Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
6
|
Zhao W, Ahmed S, Ahmed S, Yangliu Y, Wang H, Cai X. Analysis of long non-coding RNAs in epididymis of cattleyak associated with male infertility. Theriogenology 2020; 160:61-71. [PMID: 33181482 DOI: 10.1016/j.theriogenology.2020.10.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/07/2020] [Accepted: 10/26/2020] [Indexed: 02/08/2023]
Abstract
Cattleyak (CY), is a cross breed between cattle and yak (YK), which display equal adaptability to the harsh environment as YK and much higher performances than YK. However, the CY is female fertile and male sterile. Previous studies were conducted on testes tissues to investigate the mechanism of male infertility in CY. There is no systematic research on genes, especially lncRNAs between CY and YK epididymis. In this study, Illumina Hiseq was performed to profile the epididymis transcriptome (lncRNA and mRNA) of CY and YK. In total 18859 lncRNAs were identified, from which lincRNAs 12458, antisense lncRNAs 2345, intronic lncRNAs 3101, and sense lncRNAs 955 respectively. We have identified 345 DE lncRNAs and 3008 DE mRNAs between YK and CY epididymis. Thirteen DEGs were validated by quantitative real-time PCR. Combing with DEG, 14 couples of lncRNAs and their target genes were both DE, and 6 of them including CCDC39, KCNJ16, NECTIN2, MRPL20, PSMC4, and DEFB112 show their potential infertility-related terms such as cellular motility, sperm maturation, sperm storage, cellular junction, folate metabolism, and capacitation. On the other hand, several down-regulated genes such as DEFB124, DEFB126, DEFB125, DEFB127, DEFB129, CES5A, TKDP1, CST3, RNASE9 and CD52 in CY compared to YK were involved in the immune response and sperm maturation. Therefore, comprehensive analysis for lncRNAs and their target genes may enhance our understanding of the molecular mechanisms underlying the process of sperm maturation in CY and may provide important resources for further research.
Collapse
Affiliation(s)
- Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Saeed Ahmed
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Siraj Ahmed
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Yueling Yangliu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Hongmei Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization (Southwest Minzu University), Ministry of Education, Chengdu, Sichuan, 610041, China; Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
7
|
Rouaud F, Sluysmans S, Flinois A, Shah J, Vasileva E, Citi S. Scaffolding proteins of vertebrate apical junctions: structure, functions and biophysics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183399. [DOI: 10.1016/j.bbamem.2020.183399] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
|
8
|
Abstract
Epithelial cells form highly organized polarized sheets with characteristic cell morphologies and tissue architecture. Cell–cell adhesion and intercellular communication are prerequisites of such cohesive sheets of cells, and cell connectivity is mediated through several junctional assemblies, namely desmosomes, adherens, tight and gap junctions. These cell–cell junctions form signalling hubs that not only mediate cell–cell adhesion but impact on multiple aspects of cell behaviour, helping to coordinate epithelial cell shape, polarity and function. This review will focus on the tight and adherens junctions, constituents of the apical junctional complex, and aims to provide a comprehensive overview of the complex signalling that underlies junction assembly, integrity and plasticity.
Collapse
Affiliation(s)
- Alexandra D Rusu
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Marios Georgiou
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
9
|
Ugalde-Silva P, Navarro-Garcia F. Coordinated transient interaction of ZO-1 and afadin is required for pedestal maturation induced by EspF from enteropathogenic Escherichia coli. Microbiologyopen 2019; 8:e931. [PMID: 31568664 PMCID: PMC6925160 DOI: 10.1002/mbo3.931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 01/09/2023] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) infection causes a histopathological lesion including recruitment of F‐actin beneath the attached bacteria and formation of actin‐rich pedestal‐like structures. Another important target of EPEC is the tight junction (TJ), and EspF induces displacement of TJ proteins and increased intestinal permeability. Previously, we determined that an EPEC strain lacking EspF did not cause TJ disruption; meanwhile, pedestals were located on the TJ and smaller than those induced by the wild‐type strain. Therefore, EspF could be playing an important role in both phenotypes. Here, using different cell models, we found that EspF was essential for pedestal maturation through ZO‐1 disassembly from TJ, leading to (a) ZO‐1 recruitment to the pedestal structure; no other main TJ proteins were required. Recruited ZO‐1 allowed the afadin recruitment. (b) Afadin recruitment caused an afadin–ZO‐1 transient interaction, like during TJ formation. (c) Afadin and ZO‐1 were segregated to the tip and the stem of pedestal, respectively, causing pedestal maturation. Initiation of these three discrete phases for pedestal maturation functionally and physically required EspF expression. Pedestal maturation process could help coordinate the epithelial actomyosin function by maintaining the actin‐rich column composing the pedestal structure and could be important in the dynamics of the pedestal movement on epithelial cells.
Collapse
Affiliation(s)
- Paul Ugalde-Silva
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México City, Mexico
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México City, Mexico
| |
Collapse
|
10
|
Wang F, Cui Y, Shen X, Wang S, Yang GB. IL-17A and IL-17F repair HIV-1 gp140 damaged Caco-2 cell barriers by upregulating tight junction genes. Microbes Infect 2019; 21:393-400. [PMID: 30951887 DOI: 10.1016/j.micinf.2019.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/13/2019] [Accepted: 03/26/2019] [Indexed: 01/04/2023]
Abstract
It is widely accepted that impairment of the intestinal epithelial barrier from HIV/AIDS contributes significantly to microbial translocation and systemic immune activation. Such factors present potential targets for novel treatments aimed toward a functional cure. However, the extracellular mechanisms of intestinal barrier repair are poorly understood. In the current study, we investigated the abilities of IL-17A and IL-17F to repair the damaged barrier caused by HIV-1 gp140 using Caco-2 monolayers. It was found that HIV-1 gp140 downregulated the expression of tight junction-associated genes and disrupted the barrier integrity of Caco-2 monolayers. However, IL-17A and IL-17F treatment reversed the HIV-1 gp140-induced barrier dysfunction by upregulating the expression of tight junction-associated genes, the combination of which resulted in a stronger induction of barrier repair. Furthermore, the effects of IL-17A and IL-17F were reduced by downregulation of Act1 with siRNA and inhibition of NF-κB and MAPK pathways with BAY11-7082 and U0126, respectively. These data indicated that the NF-κB and MAPK pathways are involved in the repair of barrier integrity mediated by IL-17A and IL-17F, and IL-17 pathways are potential targets for gut barrier restoration therapies during HIV/AIDS.
Collapse
Affiliation(s)
- Fengjie Wang
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing, PR China
| | - Yanfang Cui
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing, PR China
| | - Xiuli Shen
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing, PR China
| | - Shuhui Wang
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing, PR China
| | - Gui-Bo Yang
- National Center for AIDS/STD Control and Prevention, China-CDC, Beijing, PR China.
| |
Collapse
|
11
|
Marques MS, Melo J, Cavadas B, Mendes N, Pereira L, Carneiro F, Figueiredo C, Leite M. Afadin Downregulation by Helicobacter pylori Induces Epithelial to Mesenchymal Transition in Gastric Cells. Front Microbiol 2018; 9:2712. [PMID: 30473688 PMCID: PMC6237830 DOI: 10.3389/fmicb.2018.02712] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 10/23/2018] [Indexed: 12/30/2022] Open
Abstract
Afadin is a cytoplasmic protein of the adherens junctions, which regulates the formation and stabilization of both the adherens and the tight junctions. Aberrant expression of Afadin has been shown in cancer and its loss has been associated with epithelial-to-mesenchymal transition (EMT). EMT is characterized by the change from an epithelial to a mesenchymal phenotype, with modifications on the expression of adhesion molecules and acquisition of a migratory and invasive cell behavior. While it is known that Helicobacter pylori disrupts the tight and the adherens junctions and induces EMT, the effect of the bacteria on Afadin is still unknown. The aim of this study was to disclose the effect of H. pylori on Afadin and its impact in the induction of an EMT phenotype in gastric cells. Using two different cell lines, we observed that H. pylori infection decreased Afadin protein levels, independently of CagA, T4SS, and VacA virulence factors. H. pylori infection of cell lines recapitulated several EMT features, displacing and downregulating multiple proteins from cell–cell junctions, and increasing the expression of ZEB1, Vimentin, Slug, N-cadherin, and Snail. Silencing of Afadin by RNAi promoted delocalization of junctional proteins from the cell–cell contacts, increased paracellular permeability, and decreased transepithelial electrical resistance, all compatible with impaired junctional integrity. Afadin silencing also led to increased expression of the EMT marker Snail, and to the formation of actin stress fibers, together with increased cell motility and invasion. Finally, and in line with our in vitro data, the gastric mucosa of individuals infected with H. pylori showed decrease/loss of Afadin membrane staining at cell–cell contacts significantly more frequently than uninfected individuals. In conclusion, Afadin is downregulated by H. pylori infection in vitro and in vivo, and its downregulation leads to the emergence of EMT and to the acquisition of an aggressive phenotype in gastric cells, which can contribute to gastric carcinogenesis.
Collapse
Affiliation(s)
- Miguel Sardinha Marques
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joana Melo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Bruno Cavadas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Nuno Mendes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Luísa Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Fátima Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Pathology, Centro Hospitalar São João, Porto, Portugal
| | - Ceu Figueiredo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Marina Leite
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
12
|
Rowart P, Wu J, Caplan MJ, Jouret F. Implications of AMPK in the Formation of Epithelial Tight Junctions. Int J Mol Sci 2018; 19:E2040. [PMID: 30011834 PMCID: PMC6073107 DOI: 10.3390/ijms19072040] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 01/13/2023] Open
Abstract
Tight junctions (TJ) play an essential role in the epithelial barrier. By definition, TJ are located at the demarcation between the apical and baso-lateral domains of the plasma membrane in epithelial cells. TJ fulfill two major roles: (i) TJ prevent the mixing of membrane components; and (ii) TJ regulate the selective paracellular permeability. Disruption of TJ is regarded as one of the earliest hallmarks of epithelial injury, leading to the loss of cell polarity and tissue disorganization. Many factors have been identified as modulators of TJ assembly/disassembly. More specifically, in addition to its role as an energy sensor, adenosine monophosphate-activated protein kinase (AMPK) participates in TJ regulation. AMPK is a ubiquitous serine/threonine kinase composed of a catalytic α-subunit complexed with regulatory β-and γ-subunits. AMPK activation promotes the early stages of epithelial TJ assembly. AMPK phosphorylates the adherens junction protein afadin and regulates its interaction with the TJ-associated protein zonula occludens (ZO)-1, thereby facilitating ZO-1 distribution to the plasma membrane. In the present review, we detail the signaling pathways up-and down-stream of AMPK activation at the time of Ca2+-induced TJ assembly.
Collapse
Affiliation(s)
- Pascal Rowart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Avenue de L'Hôpital 11, 4000 Liège, Belgium.
| | - Jingshing Wu
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - François Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège (ULiège), Avenue de L'Hôpital 11, 4000 Liège, Belgium.
- Division of Nephrology, Centre Hospitalier Universitaire de Liège (CHU of Liège), University of Liège (CHU ULiège), 13-B4000 Liège, Belgium.
| |
Collapse
|
13
|
Oshima T, Miyashita H, Ishimura Y, Ito Y, Tanaka Y, Hori A, Kokubo T, Kurokawa T. Fc engineering of anti-Nectin-2 antibody improved thrombocytopenic adverse event in monkey. PLoS One 2018; 13:e0196422. [PMID: 29723247 PMCID: PMC5933732 DOI: 10.1371/journal.pone.0196422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 04/12/2018] [Indexed: 11/18/2022] Open
Abstract
Nectin-2 is a transmembrane glycoprotein which is involved in the process of Ca2+-independent cell-cell adhesion. In our previous study, we have demonstrated that Nectin-2 is over-expressed in breast and ovarian cancer tissues by using gene expression analysis and immunohistochemistry. Furthermore, we discovered multiple anti-Nectin-2 fully human monoclonal antibodies which inhibited tumor growth in in vivo subcutaneous xenograft models with antibody-dependent cellular cytotoxicity (ADCC) as the principal mechanism of action. In this report, we assessed the toxicity of Y-443, a fully human IgG1/kappa anti-Nectin-2 monoclonal antibody exhibiting strong in vitro ADCC and in vivo anti-tumor activity in cynomolgus monkeys (Macaca fascicularis (Cynos)). Unexpectedly, upon administration, Y-443 induced strong thrombocytopenia through Nectin-2 expressed on Cyno platelets, presumably followed by phagocytosis in the mononuclear phagocytic system. To mitigate the adverse safety profile, we mutated the Fc region of Y-443 to reduce the Fc binding activity to Fcγ receptor I, which is the primary receptor for phagocytosis on macrophages. Moreover, we further engineered the Fc through defucosylation to maintain ADCC activity. The resultant Fc engineered antibody, termed Y-634, demonstrated diminished thrombocytopenia in Cyno toxicological studies and maintained anti-tumor activity in a mouse xenograft model. These findings suggest that Y-634 may have a therapeutic potential for the treatment of Nectin-2 positive cancers, and moreover, Fc engineering is a potential mitigation strategy to ameliorate safety liabilities in antibody induced thrombocytopenia while maintaining antibody potency.
Collapse
Affiliation(s)
- Tsutomu Oshima
- Immunobiologics, Takeda California Inc., San Diego, California, United States of America
- * E-mail:
| | - Hideaki Miyashita
- Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, Hikari, Yamaguchi, Japan
| | - Yoshimasa Ishimura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Yuki Ito
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Yoko Tanaka
- Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, Hikari, Yamaguchi, Japan
| | - Akira Hori
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Toshio Kokubo
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Tomofumi Kurokawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
14
|
Tsou PS, Wren JD, Amin MA, Schiopu E, Fox DA, Khanna D, Sawalha AH. Histone Deacetylase 5 Is Overexpressed in Scleroderma Endothelial Cells and Impairs Angiogenesis via Repression of Proangiogenic Factors. Arthritis Rheumatol 2017; 68:2975-2985. [PMID: 27482699 DOI: 10.1002/art.39828] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Vascular dysfunction represents a disease-initiating event in systemic sclerosis (SSc; scleroderma). Results of recent studies suggest that epigenetic dysregulation impairs normal angiogenesis and can result in abnormal patterns of blood vessel growth. Histone deacetylases (HDACs) control endothelial cell (EC) proliferation and regulate EC migration. Specifically, HDAC-5 appears to be antiangiogenic. This study was undertaken to test whether HDAC-5 contributes to impaired angiogenesis in SSc by repressing proangiogenic factors in ECs. METHODS Dermal ECs were isolated from patients with diffuse cutaneous SSc and healthy controls. Angiogenesis was assessed using an in vitro Matrigel tube formation assay. An assay for transposase-accessible chromatin using sequencing (ATAC-seq) was performed to assess and localize the genome-wide effects of HDAC5 knockdown on chromatin accessibility. RESULTS The expression of HDAC5 was significantly increased in ECs from patients with SSc compared to healthy control ECs. Silencing of HDAC5 in SSc ECs restored normal angiogenesis. HDAC5 knockdown followed by ATAC-seq assay in SSc ECs identified key HDAC5-regulated genes involved in angiogenesis and fibrosis, such as CYR61, PVRL2, and FSTL1. Simultaneous knockdown of HDAC5 in conjunction with either CYR61, PVRL2, or FSTL1 inhibited angiogenesis in SSc ECs. Conversely, overexpression of these genes individually led to an increase in tube formation as assessed by Matrigel assay, suggesting that these genes play functional roles in the impairment of angiogenesis in SSc. CONCLUSION Several novel HDAC5-regulated target genes associated with impaired angiogenesis were identified in SSc ECs by ATAC-seq. The results of this study provide a potential link between epigenetic regulation and impaired angiogenesis in SSc, and identify a novel mechanism for the dysregulated angiogenesis that characterizes this disease.
Collapse
Affiliation(s)
| | - Jonathan D Wren
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City
| | | | | | | | | | | |
Collapse
|
15
|
Zihni C, Mills C, Matter K, Balda MS. Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol 2016; 17:564-80. [PMID: 27353478 DOI: 10.1038/nrm.2016.80] [Citation(s) in RCA: 997] [Impact Index Per Article: 110.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epithelia and endothelia separate different tissue compartments and protect multicellular organisms from the outside world. This requires the formation of tight junctions, selective gates that control paracellular diffusion of ions and solutes. Tight junctions also form the border between the apical and basolateral plasma-membrane domains and are linked to the machinery that controls apicobasal polarization. Additionally, signalling networks that guide diverse cell behaviours and functions are connected to tight junctions, transmitting information to and from the cytoskeleton, nucleus and different cell adhesion complexes. Recent advances have broadened our understanding of the molecular architecture and cellular functions of tight junctions.
Collapse
Affiliation(s)
- Ceniz Zihni
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Clare Mills
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Karl Matter
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Maria S Balda
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| |
Collapse
|
16
|
Karabulut M, Gunaldi M, Alis H, Afsar CU, Karabulut S, Serilmez M, Akarsu C, Seyit H, Aykan NF. Serum nectin-2 levels are diagnostic and prognostic in patients with colorectal carcinoma. Clin Transl Oncol 2016; 18:160-171. [PMID: 26184725 DOI: 10.1007/s12094-015-1348-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/02/2015] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Nectins are a family of integral protein and immunoglobulin-like cell adhesion molecules involved in the formation of functioning adherence and tight junctions. Aberrant expression is associated with cancer progression, apoptosis and cell proliferation but little is known how these effects change in cell behavior. The objective of this study was to evaluate the serum levels of nectin-2 with regard to diagnostic, predictive and prognostic value in colorectal cancer (CRC) patients. MATERIALS AND METHODS One-hundred and forty CRC patients were enrolled in this study. Serum nectin-2 levels were determined by enzyme-linked immunosorbent assay method. Age- and sex-matched 40 healthy controls were included in the analysis. RESULTS Median age of patients was 60 years old, range 24-84 years. The localization of tumor in majority of the patients was colon (n = 81, 58 %). Non-metastatic (stage II and III) and metastatic patients' baseline serum nectin-2 levels were significantly higher than those in the healthy control group (p < 0.001; for two group). However, known clinical variables including response to CTx (chemotherapy) were not found to be correlated with serum nectin-2 concentrations (p > 0.05). While non-metastatic group patients with elevated serum nectin-2 levels showed significant adverse effect on PFS, metastatic group patients with elevated serum nectin-2 levels showed no significant adverse effect on PFS (p = 0.05 and p = 0.29, respectively). On the other hand, our study results did not show statistically significant serum nectin-2 concentrations regarding overall survival rates. CONCLUSION Serum levels of nectin-2 may have diagnostic roles for CRC patients. Moreover, our study results show the prognostic role of nectin-2 in non-metastatic group patients.
Collapse
Affiliation(s)
- M Karabulut
- Clinic of General Surgery, Istanbul Bakırköy Dr. Sadi Konuk Education and Research Hospital, Istanbul, Turkey
| | - M Gunaldi
- Clinic of Medical Oncology, Istanbul Bakirkoy Dr. Sadi Konuk Education and Research Hospital, Istanbul, Turkey
| | - H Alis
- Clinic of General Surgery, Istanbul Bakırköy Dr. Sadi Konuk Education and Research Hospital, Istanbul, Turkey
| | - C U Afsar
- Department of Medical Oncology, Ministry of Health, Istanbul Training and Research Hospital, Istanbul, Turkey.
| | - S Karabulut
- Department of Medical Oncology, Institute of Oncology, Istanbul University, Istanbul, Turkey
| | - M Serilmez
- Department of Basic Oncology, Institute of Oncology, Istanbul University, Istanbul, Turkey
| | - C Akarsu
- Clinic of General Surgery, Istanbul Bakırköy Dr. Sadi Konuk Education and Research Hospital, Istanbul, Turkey
| | - H Seyit
- Clinic of General Surgery, Istanbul Bakırköy Dr. Sadi Konuk Education and Research Hospital, Istanbul, Turkey
| | - N F Aykan
- Department of Medical Oncology, Institute of Oncology, Istanbul University, Istanbul, Turkey
| |
Collapse
|
17
|
Jheon AH, Prochazkova M, Meng B, Wen T, Lim YJ, Naveau A, Espinoza R, Sone ED, Ganss B, Siebel CW, Klein OD. Inhibition of Notch Signaling During Mouse Incisor Renewal Leads to Enamel Defects. J Bone Miner Res 2016; 31:152-62. [PMID: 26179131 PMCID: PMC4840178 DOI: 10.1002/jbmr.2591] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 06/18/2015] [Accepted: 07/02/2015] [Indexed: 12/13/2022]
Abstract
The continuously growing rodent incisor is an emerging model for the study of renewal of mineralized tissues by adult stem cells. Although the Bmp, Fgf, Shh, and Wnt pathways have been studied in this organ previously, relatively little is known about the role of Notch signaling during incisor renewal. Notch signaling components are expressed in enamel-forming ameloblasts and the underlying stratum intermedium (SI), which suggested distinct roles in incisor renewal and enamel mineralization. Here, we injected adult mice with inhibitory antibodies against several components of the Notch pathway. This blockade led to defects in the interaction between ameloblasts and the SI cells, which ultimately affected enamel formation. Furthermore, Notch signaling inhibition led to the downregulation of desmosome-specific proteins such as PERP and desmoplakin, consistent with the importance of desmosomes in the integrity of ameloblast-SI attachment and enamel formation. Together, our data demonstrate that Notch signaling is critical for proper enamel formation during incisor renewal, in part by regulating desmosome-specific components, and that the mouse incisor provides a model system to dissect Jag-Notch signaling mechanisms in the context of mineralized tissue renewal.
Collapse
Affiliation(s)
- Andrew H. Jheon
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
| | - Michaela Prochazkova
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
- Department of Anthropology and Human Genetics, Charles University in Prague, Czech Republic
| | - Bo Meng
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
| | - Timothy Wen
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
| | - Young-Jun Lim
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
- Seoul National University, Seoul, South Korea
| | - Adrien Naveau
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
| | - Ruben Espinoza
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
| | - Eli D. Sone
- Institute of Biomaterials and Biomedical Engineering, Department of Materials Science and Engineering, and Faculty of Dentistry, University of Toronto, Ontario, Canada
| | - Bernhard Ganss
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Ontario, Canada
| | | | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, USA
- Department of Pediatrics and Institute for Human Genetics, University of California San Francisco, San Francisco, USA
| |
Collapse
|
18
|
Bauer HC, Krizbai IA, Bauer H, Traweger A. "You Shall Not Pass"-tight junctions of the blood brain barrier. Front Neurosci 2014; 8:392. [PMID: 25520612 PMCID: PMC4253952 DOI: 10.3389/fnins.2014.00392] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/14/2014] [Indexed: 12/31/2022] Open
Abstract
The structure and function of the barrier layers restricting the free diffusion of substances between the central nervous system (brain and spinal cord) and the systemic circulation is of great medical interest as various pathological conditions often lead to their impairment. Excessive leakage of blood-borne molecules into the parenchyma and the concomitant fluctuations in the microenvironment following a transient breakdown of the blood-brain barrier (BBB) during ischemic/hypoxic conditions or because of an autoimmune disease are detrimental to the physiological functioning of nervous tissue. On the other hand, the treatment of neurological disorders is often hampered as only minimal amounts of therapeutic agents are able to penetrate a fully functional BBB or blood cerebrospinal fluid barrier. An in-depth understanding of the molecular machinery governing the establishment and maintenance of these barriers is necessary to develop rational strategies allowing a controlled delivery of appropriate drugs to the CNS. At the basis of such tissue barriers are intimate cell-cell contacts (zonulae occludentes, tight junctions) which are present in all polarized epithelia and endothelia. By creating a paracellular diffusion constraint TJs enable the vectorial transport across cell monolayers. More recent findings indicate that functional barriers are already established during development, protecting the fetal brain. As an understanding of the biogenesis of TJs might reveal the underlying mechanisms of barrier formation during ontogenic development numerous in vitro systems have been developed to study the assembly and disassembly of TJs. In addition, monitoring the stage-specific expression of TJ-associated proteins during development has brought much insight into the “developmental tightening” of tissue barriers. Over the last two decades a detailed molecular map of transmembrane and cytoplasmic TJ-proteins has been identified. These proteins not only form a cell-cell adhesion structure, but integrate various signaling pathways, thereby directly or indirectly impacting upon processes such as cell-cell adhesion, cytoskeletal rearrangement, and transcriptional control. This review will provide a brief overview on the establishment of the BBB during embryonic development in mammals and a detailed description of the ultrastructure, biogenesis, and molecular composition of epithelial and endothelial TJs will be given.
Collapse
Affiliation(s)
- Hans-Christian Bauer
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University - Spinal Cord Injury and Tissue Regeneration Center Salzburg Salzburg, Austria ; Department of Traumatology and Sports Injuries, Paracelsus Medical University Salzburg, Austria ; Austrian Cluster for Tissue Regeneration Vienna, Austria
| | - István A Krizbai
- Biological Research Centre, Institute of Biophysics, Hungarian Academy of Sciences Szeged, Hungary ; Institute of Life Sciences, Vasile Goldis Western University of Arad Arad, Romania
| | - Hannelore Bauer
- Department of Organismic Biology, University of Salzburg Salzburg, Austria
| | - Andreas Traweger
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University - Spinal Cord Injury and Tissue Regeneration Center Salzburg Salzburg, Austria ; Austrian Cluster for Tissue Regeneration Vienna, Austria
| |
Collapse
|
19
|
Zhang X, Lui WY. Dysregulation of nectin-2 in the testicular cells: an explanation of cadmium-induced male infertility. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:873-84. [PMID: 25046863 DOI: 10.1016/j.bbagrm.2014.07.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/19/2014] [Accepted: 07/14/2014] [Indexed: 12/14/2022]
Abstract
Nectin-2, a junction molecule, is found at the basal and apical ectoplasmic specializations (ES) for the formation of the blood-testis barrier (BTB) (constituted by tight junctions and basal ES) and Sertoli-spermatid adhesion. Loss of nectin-2 causes male infertility, suggesting nectin-2-based ES is crucial for spermatogenesis. Cadmium (Cd) has been known to induce severe testicular injury. Recent evidence has shown that the basal ES at the BTB and apical ES are the targets of Cd, suggesting that unique junction protein at the ES may explain why testis is more susceptible than other tissues. Since nectin-2 is expressed exclusively at the ES, it is highly possible that nectin-2 is the direct target of Cd. In this study, we investigate if nectin-2 is the target protein of Cd toxicity and the mechanism on how Cd down-regulates nectin-2 to achieve ES disruption. Our results revealed that Cd suppresses nectin-2 at transcriptional and post-translational levels. Inhibitor and shRNA knockdown have shown that Cd induces nectin-2 protein degradation via clathrin-dependent endocytosis. Immunofluorescence staining and endocytosis assays further confirmed that nectin-2 internalization is promoted upon Cd treatment. Besides, Cd directly represses nectin-2 transcription. EMSA and ChIP assays showed that Cd inhibits the binding of positive regulators to nectin-2 promoter. siRNA and overexpression analyses have demonstrated that Cd reduces the expression and binding affinity of positive regulators for transcription. Taken together, nectin-2 is the direct molecular target of Cd and its disruptive effects are mediated via direct repressing nectin-2 transcription and endocytosis of nectin-2 for degradation.
Collapse
Affiliation(s)
- Xu Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Wing-Yee Lui
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
20
|
Yamamoto H, Maruo T, Majima T, Ishizaki H, Tanaka-Okamoto M, Miyoshi J, Mandai K, Takai Y. Genetic deletion of afadin causes hydrocephalus by destruction of adherens junctions in radial glial and ependymal cells in the midbrain. PLoS One 2013; 8:e80356. [PMID: 24236178 PMCID: PMC3827428 DOI: 10.1371/journal.pone.0080356] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/11/2013] [Indexed: 12/23/2022] Open
Abstract
Adherens junctions (AJs) play a role in mechanically connecting adjacent cells to maintain tissue structure, particularly in epithelial cells. The major cell–cell adhesion molecules at AJs are cadherins and nectins. Afadin binds to both nectins and α-catenin and recruits the cadherin-β-catenin complex to the nectin-based cell–cell adhesion site to form AJs. To explore the role of afadin in radial glial and ependymal cells in the brain, we generated mice carrying a nestin-Cre-mediated conditional knockout (cKO) of the afadin gene. Newborn afadin-cKO mice developed hydrocephalus and died neonatally. The afadin-cKO brain displayed enlarged lateral ventricles and cerebral aqueduct, resulting from stenosis of the caudal end of the cerebral aqueduct and obliteration of the ventral part of the third ventricle. Afadin deficiency further caused the loss of ependymal cells from the ventricular and aqueductal surfaces. During development, radial glial cells, which terminally differentiate into ependymal cells, scattered from the ventricular zone and were replaced by neurons that eventually covered the ventricular and aqueductal surfaces of the afadin-cKO midbrain. Moreover, the denuded ependymal cells were only occasionally observed in the third ventricle and the cerebral aqueduct of the afadin-cKO midbrain. Afadin was co-localized with nectin-1 and N-cadherin at AJs of radial glial and ependymal cells in the control midbrain, but these proteins were not concentrated at AJs in the afadin-cKO midbrain. Thus, the defects in the afadin-cKO midbrain most likely resulted from the destruction of AJs, because AJs in the midbrain were already established before afadin was genetically deleted. These results indicate that afadin is essential for the maintenance of AJs in radial glial and ependymal cells in the midbrain and is required for normal morphogenesis of the cerebral aqueduct and ventral third ventricle in the midbrain.
Collapse
Affiliation(s)
- Hideaki Yamamoto
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Tomohiko Maruo
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Takashi Majima
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
| | - Hiroyoshi Ishizaki
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
| | - Miki Tanaka-Okamoto
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
| | - Jun Miyoshi
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
| | - Kenji Mandai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- * E-mail: (KT); (KM)
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- * E-mail: (KT); (KM)
| |
Collapse
|
21
|
Yamada T, Kuramitsu K, Rikitsu E, Kurita S, Ikeda W, Takai Y. Nectin and junctional adhesion molecule are critical cell adhesion molecules for the apico-basal alignment of adherens and tight junctions in epithelial cells. Genes Cells 2013; 18:985-98. [PMID: 24112238 DOI: 10.1111/gtc.12091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/30/2013] [Indexed: 02/02/2023]
Abstract
Tight junctions (TJs) and adherens junctions (AJs) form an apical junctional complex at the apical side of the lateral membranes of epithelial cells, in which TJs are aligned at the apical side of AJs. Many cell adhesion molecules (CAMs) and cell polarity molecules (CPMs) cooperatively regulate the formation of the apical junctional complex, but the mechanism for the alignment of TJs at the apical side of AJs is not fully understood. We developed a cellular system with which epithelial-like TJs and AJs were reconstituted in fibroblasts and analyzed the cooperative roles of CAMs and CPMs. We exogenously expressed various combinations of CAMs and CPMs in fibroblasts that express negligible amounts of these molecules endogenously. In these cells, the nectin-based cell-cell adhesion was formed at the apical side of the junctional adhesion molecule (JAM)-based cell-cell adhesion, and cadherin and claudin were recruited to the nectin-3- and JAM-based cell-cell adhesion sites to form AJ-like and TJ-like domains, respectively. This inversed alignment of the AJ-like and TJ-like domains was reversed by complementary expression of CPMs Par-3, atypical protein kinase C, Par-6, Crb3, Pals1 and Patj. We describe the cooperative roles of these CAMs and CPMs in the apico-basal alignment of TJs and AJs in epithelial cells.
Collapse
Affiliation(s)
- Tomohiro Yamada
- KAN Research Institute, Inc., 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Mok KW, Mruk DD, Cheng CY. Regulation of blood-testis barrier (BTB) dynamics during spermatogenesis via the "Yin" and "Yang" effects of mammalian target of rapamycin complex 1 (mTORC1) and mTORC2. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:291-358. [PMID: 23317821 DOI: 10.1016/b978-0-12-407704-1.00006-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In mammalian testes, haploid spermatozoa are formed from diploid spermatogonia during spermatogenesis, which is a complicated cellular process. While these cellular events were reported in the 1960s and 1970s, the underlying molecular mechanism(s) that regulates these events remained unexplored until the past ∼10 years. For instance, adhesion proteins were shown to be integrated components at the Sertoli cell-cell interface and/or the Sertoli-spermatid interface in the late 1980s. But only until recently, studies have demonstrated that some of the adhesion proteins serve as the platform for signal transduction that regulates cell adhesion. In this chapter, a brief summary and critical discussion are provided on the latest findings regarding these cell-adhesion proteins in the testis and their relationship to spermatogenesis. Moreover, antagonistic effects of two mammalian target of rapamycin (mTOR) complexes, known as mTORC1 and mTORC2, on cell-adhesion function in the testis are discussed. Finally, a hypothetic model is presented to depict how these two mTOR-signaling complexes having the "yin" and "yang" antagonistic effects on the Sertoli cell tight junction (TJ)-permeability barrier can maintain the blood-testis barrier (BTB) integrity during the epithelial cycle while preleptotene spermatocytes are crossing the BTB.
Collapse
Affiliation(s)
- Ka Wai Mok
- Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, USA
| | | | | |
Collapse
|
23
|
Oshima T, Sato S, Kato J, Ito Y, Watanabe T, Tsuji I, Hori A, Kurokawa T, Kokubo T. Nectin-2 is a potential target for antibody therapy of breast and ovarian cancers. Mol Cancer 2013; 12:60. [PMID: 23758976 PMCID: PMC3698035 DOI: 10.1186/1476-4598-12-60] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/31/2013] [Indexed: 01/22/2023] Open
Abstract
Background Nectin-2 is a Ca2+-independent cell-cell adhesion molecule that is one of the plasma membrane components of adherens junctions. However, little has been reported about the involvement of Nectin-2 in cancer. Methods To determine the expression of Nectin-2 in cancer tissues and cancer cell lines, we performed gene expression profile analysis, immunohistochemistry studies, and flow cytometry analysis. We also investigated the potential of this molecule as a target for antibody therapeutics to treat cancers by generating and characterizing an anti-Nectin-2 rabbit polyclonal antibody (poAb) and 256 fully human anti-Nectin-2 monoclonal antibodies (mAbs). In addition, we tested anti-Nectin-2 mAbs in several in vivo tumor growth inhibition models to investigate the primary mechanisms of action of the mAbs. Results In the present study, we found that Nectin-2 was over-expressed in clinical breast and ovarian cancer tissues by using gene expression profile analysis and immunohistochemistry studies. Nectin-2 was over-expressed in various cancer cell lines as well. Furthermore, the polyclonal antibody specific to Nectin-2 suppressed the in vitro proliferation of OV-90 ovarian cancer cells, which express endogenous Nectin-2 on the cell surface. The anti-Nectin-2 mAbs we generated were classified into 7 epitope bins. The anti-Nectin-2 mAbs demonstrated antibody-dependent cellular cytotoxicity (ADCC) and epitope bin-dependent features such as the inhibition of Nectin-2-Nectin-2 interaction, Nectin-2-Nectin-3 interaction, and in vitro cancer cell proliferation. A representative anti-Nectin-2 mAb in epitope bin VII, Y-443, showed anti-tumor effects against OV-90 cells and MDA-MB-231 breast cancer cells in mouse therapeutic models, and its main mechanism of action appeared to be ADCC. Conclusions We observed the over-expression of Nectin-2 in breast and ovarian cancers and anti-tumor activity of anti-Nectin-2 mAbs via strong ADCC. These findings suggest that Nectin-2 is a potential target for antibody therapy against breast and ovarian cancers.
Collapse
Affiliation(s)
- Tsutomu Oshima
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraokahigashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Rikitake Y, Mandai K, Takai Y. The role of nectins in different types of cell-cell adhesion. J Cell Sci 2013; 125:3713-22. [PMID: 23027581 DOI: 10.1242/jcs.099572] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mammalian tissues and organs are composed of different types of cells that adhere to each other homotypically (i.e. interactions between cells of the same cell type) or heterotypically (i.e. interactions between different cell types), forming a variety of cellular patterns, including mosaic patterns. At least three types of cell-cell adhesion have been observed: symmetric homotypic, asymmetric homotypic and heterotypic cell adhesions. Cadherins and nectins, which are known cell-cell adhesion molecules, mediate these cell adhesions. Cadherins comprise a family of more than 100 members, but they are primarily involved in homophilic trans-interactions (i.e. interactions between the same cadherin members) between opposing cells. By contrast, the nectin family comprises only four members, and these proteins form both homophilic and heterophilic trans-interactions (i.e. interactions between the same and different nectin members on opposing cells). In addition, heterophilic trans-interactions between nectins are much stronger than homophilic trans-interactions. Because of these unique properties, nectins have crucial roles in asymmetric homotypic cell-cell adhesion at neuronal synapses and in various types of heterotypic cell-cell adhesions. We summarize recent progress in our understanding of the biology of nectins and discuss their roles in heterotypic cell-cell adhesions, whose formation cannot be solely explained by the action of cadherins.
Collapse
Affiliation(s)
- Yoshiyuki Rikitake
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | | | | |
Collapse
|
25
|
Huang RYJ, Guilford P, Thiery JP. Early events in cell adhesion and polarity during epithelial-mesenchymal transition. J Cell Sci 2013; 125:4417-22. [PMID: 23165231 DOI: 10.1242/jcs.099697] [Citation(s) in RCA: 259] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Ruby Yun-Ju Huang
- Department of Obstetrics and Gynaecology, National University Hospital, 119074, Singapore
| | | | | |
Collapse
|
26
|
Yang Z, Zimmerman S, Brakeman PR, Beaudoin GM, Reichardt LF, Marciano DK. De novo lumen formation and elongation in the developing nephron: a central role for afadin in apical polarity. Development 2013; 140:1774-84. [PMID: 23487309 DOI: 10.1242/dev.087957] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A fundamental process in biology is the de novo formation and morphogenesis of polarized tubules. Although these processes are essential for the formation of multiple metazoan organ systems, little is known about the molecular mechanisms that regulate them. In this study, we have characterized several steps in tubule formation and morphogenesis using the mouse kidney as a model system. We report that kidney mesenchymal cells contain discrete Par3-expressing membrane microdomains that become restricted to an apical domain, coinciding with lumen formation. Once lumen formation has been initiated, elongation occurs by simultaneous extension and additional de novo lumen generation. We demonstrate that lumen formation and elongation require afadin, a nectin adaptor protein implicated in adherens junction formation. Mice that lack afadin in nephron precursors show evidence of Par3-expressing membrane microdomains, but fail to develop normal apical-basal polarity and generate a continuous lumen. Absence of afadin led to delayed and diminished integration of nectin complexes and failure to recruit R-cadherin. Furthermore, we demonstrate that afadin is required for Par complex formation. Together, these results suggest that afadin acts upstream of the Par complex to regulate the integration and/or coalescence of membrane microdomains, thereby establishing apical-basal polarity and lumen formation/elongation during kidney tubulogenesis.
Collapse
Affiliation(s)
- Zhufeng Yang
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
27
|
Dubé É, Cyr DG. The Blood-Epididymis Barrier and Human Male Fertility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 763:218-36. [DOI: 10.1007/978-1-4614-4711-5_11] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
28
|
Yamaki T, Ohtake K, Ichikawa K, Uchida M, Uchida H, Ohshima S, Juni K, Kobayashi J, Morimoto Y, Natsume H. Poly- L-arginine-Induced Internalization of Tight Junction Proteins Increases the Paracellular Permeability of the Caco-2 Cell Monolayer to Hydrophilic Macromolecules. Biol Pharm Bull 2013; 36:432-41. [DOI: 10.1248/bpb.b12-00878] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Kazuo Ohtake
- Faculty of Pharmaceutical Sciences, Josai University
| | | | - Masaki Uchida
- Faculty of Pharmaceutical Sciences, Josai University
| | | | | | - Kazuhiko Juni
- Faculty of Pharmaceutical Sciences, Josai University
| | - Jun Kobayashi
- Faculty of Pharmaceutical Sciences, Josai University
| | - Yasunori Morimoto
- Faculty of Pharmaceutical Sciences, Josai University
- Research Institute of TTS Technology, Josai University
| | - Hideshi Natsume
- Faculty of Pharmaceutical Sciences, Josai University
- Research Institute of TTS Technology, Josai University
| |
Collapse
|
29
|
Mandai K, Rikitake Y, Shimono Y, Takai Y. Afadin/AF-6 and Canoe. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:433-54. [DOI: 10.1016/b978-0-12-394311-8.00019-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Irie K, Shimizu K, Sakisaka T, Ikeda W, Takai Y. Roles of nectins in cell adhesion, signaling and polarization. Handb Exp Pharmacol 2012:343-72. [PMID: 20455098 DOI: 10.1007/978-3-540-68170-0_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Nectins are Ca(2+)-independent immunoglobulin-like cell-cell adhesion molecules which constitute a family of four members. Nectins homophilically and heterophilically trans-interact and cause cell-cell adhesion. This nectin-based cell-cell adhesion plays roles in the organization of adherens junctions in epithelial cells and fibroblasts and synaptic junctions in neurons in cooperation with cadherins. The nectin-based cell-cell adhesion plays roles in the contacts between commissural axons and floor plate cells and in the organization of Sertoli cell-spermatid junctions in the testis, independently of cadherins. Nectins furthermore regulate intracellular signaling through Cdc42 and Rac small G proteins and cell polarization through cell polarity proteins. Pathologically, nectins serve as entry and cell-cell spread mediators of herpes simplex viruses.
Collapse
Affiliation(s)
- K Irie
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine/Faculty of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | | | | | | | | |
Collapse
|
31
|
Solecki DJ. Sticky situations: recent advances in control of cell adhesion during neuronal migration. Curr Opin Neurobiol 2012; 22:791-8. [PMID: 22560352 DOI: 10.1016/j.conb.2012.04.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 03/26/2012] [Accepted: 04/12/2012] [Indexed: 11/30/2022]
Abstract
The migration of neurons along glial fibers from a germinal zone (GZ) to their final laminar positions is essential for morphogenesis of the developing brain; aberrations in this process are linked to profound neurodevelopmental and cognitive disorders. During this critical morphogenic movement, neurons must navigate complex migration paths, propelling their cell bodies through the dense cellular environment of the developing nervous system to their final destinations. It is not understood how neurons can successfully migrate along their glial guides through the myriad processes and cell bodies of neighboring neurons. Although much progress has been made in understanding the substrates (Fishell G, Hatten ME: Astrotactin provides a receptor system for CNS neuronal migration. Development 1991, 113:755; Elias LA, Wang DD, Kriegstein AR: Gap junction adhesion is necessary for radial migration in the neocortex. Nature 2007, 448:901; Anton ES, Kreidberg JA, Rakic P: Distinct functions of alpha3 and alpha. (v) integrin receptors in neuronal migration and laminar organization of the cerebral cortex. Neuron 1999, 22:277; Anton ES, Marchionni MA, Lee KF, Rakic P: Role of GGF/neuregulin signaling in interactions between migrating neurons and radial glia in the developing cerebral cortex. Development 1997, 124:3501), guidance mechanisms (Polleux F, Whitford KL, Dijkhuizen PA, Vitalis T, Ghosh A: Control of cortical interneuron migration by neurotrophins and PI3-kinase signaling. Development 2002, 129:3147; Zhou P, et al.: Polarized signaling endosomes coordinate BDNF-induced chemotaxis of cerebellar precursors. Neuron 2007, 55:53; Renaud J, et al.: Plexin-A2 and its ligand, Sema6A, control nucleus-centrosome coupling in migrating granule cells. Nat Neurosci 2008, 11:440), cytoskeletal elements (Schaar BT, McConnell SK: Cytoskeletal coordination during neuronal migration. Proc Natl Acad Sci U S A 2005, 102:13652; Tsai JW, Bremner KH, Vallee RB: Dual subcellular roles for LIS1 and dynein in radial neuronal migration in live brain tissue. Nat Neurosci 2007, 10:970; Solecki DJ, et al.: Myosin II motors and F-actin dynamics drive the coordinated movement of the centrosome and soma during CNS glial-guided neuronal migration. Neuron 2009, 63:63), and post-translational modifications (Patrick GN, Zhou P, Kwon YT, Howley PM, Tsai LH: p35, the neuronal-specific activator of cyclin-dependent kinase 5 (Cdk5) is degraded by the ubiquitin-proteasome pathway. J Biol Chem 1998, 273:24057; Suetsugu S, et al.: Regulation of actin cytoskeleton by mDab1 through N-WASP and ubiquitination of mDab1. Biochem J 2004, 384:1; Karakuzu O, Wang DP, Cameron S: MIG-32 and SPAT-3A are PRC1 homologs that control neuronal migration inCaenorhabditis elegans. Development 2009, 136:943) required for neuronal migration, we have yet to elucidate how neurons regulate their cellular interactions and adhesive specificity to follow the appropriate migratory pathways. Here I will examine recent developments in our understanding of the mechanisms controlling neuronal cell adhesion and how these mechanisms interact with crucial neurodevelopmental events, such as GZ exit, migration pathway selection, multipolar-to-radial transition, and final lamination.
Collapse
Affiliation(s)
- David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| |
Collapse
|
32
|
Abstract
Junctional adhesion molecules are transmembrane proteins that belong to the immunoglobulin superfamily. In addition to their localization in close proximity to the tight junctions in endothelial and epithelial cells, junctional adhesion molecules are also expressed in circulating cells that do not form junctions, such as leukocytes and platelets. As a consequence, these proteins are associated not only with the permeability-regulating barrier function of the tight junctions, but also with other biologic processes, such as inflammatory reactions, responses to vascular injury, and tumor angiogenesis. Furthermore, because of their transmembrane topology, junctional adhesion molecules are poised both for receiving inputs from the cell interior (their expression, localization, and function being regulated in response to inflammatory cytokines and growth factors) and for translating extracellular adhesive events into functional responses. This review focuses on the different roles of junctional adhesion molecules in normal and pathologic conditions, with emphasis on inflammatory reactions and vascular responses to injury.
Collapse
Affiliation(s)
- Gianfranco Bazzoni
- Department of Biochemistry and Molecular Pharmacology Mario Negri Institute of Pharmacological Research, Milano, Italy.
| |
Collapse
|
33
|
Dudak A, Kim J, Cheong B, Federoff HJ, Lim ST. Membrane palmitoylated proteins regulate trafficking and processing of nectins. Eur J Cell Biol 2011; 90:365-75. [PMID: 21371776 PMCID: PMC3060411 DOI: 10.1016/j.ejcb.2011.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 01/07/2011] [Accepted: 01/07/2011] [Indexed: 11/25/2022] Open
Abstract
Nectins are cell-cell adhesion molecules involved in the formation of various intercellular junctions and the establishment of apical-basal polarity at cell-cell adhesion sites. To have a better understanding of the roles of nectins in the formation of cell-cell junctions, we searched for new cytoplasmic binding partners for nectin. We report that nectin-1α associates with membrane palmitoylated protein 3 (MPP3), one of the human homologues of a Drosophila tumor suppressor gene, Disc large. Two major forms of MPP3 at 66 and 98 kDa were detected, in conjunction with nectin-1α, suggesting that an association between the two may occur in various cell types. Nectin-1α recruits MPP3 to cell-cell contact sites, mediated by a PDZ-binding motif at the carboxyl terminus of nectin-1α. Association with MPP3 increases cell surface expression of nectin-1α and enhances nectin-1α ectodomain shedding, indicating that MPP3 regulates trafficking and processing of nectin-1α. Further study showed that MPP3 interacts with nectin-3α, but not with nectin-2α, showing that the association of nectins with MPP3 is isoform-specific. MPP5, another MPP family member, interacts with nectins with varying affinity and facilitates surface expression of nectin-1α, nectin-2α, and nectin-3α. These data suggest that wide interactions between nectins and MPP family members may occur in various cell-cell junctions and that these associations may regulate trafficking and processing of nectins.
Collapse
Affiliation(s)
- Amanda Dudak
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd. NW, Washington, DC 20057
| | - Jinsook Kim
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd. NW, Washington, DC 20057
| | - Bryan Cheong
- Thomas Jefferson High School for Science and Technology, 6560 Braddock Rd., Alexandria VA, 22312
| | - Howard J. Federoff
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd. NW, Washington, DC 20057
- Department of Neurology, Georgetown University Medical Center, 3970 Reservoir Rd. NW, Washington, DC 20057
| | - Seung T. Lim
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd. NW, Washington, DC 20057
| |
Collapse
|
34
|
Jheon AH, Mostowfi P, Snead ML, Ihrie RA, Sone E, Pramparo T, Attardi LD, Klein OD. PERP regulates enamel formation via effects on cell-cell adhesion and gene expression. J Cell Sci 2011; 124:745-54. [PMID: 21285247 PMCID: PMC3039019 DOI: 10.1242/jcs.078071] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2010] [Indexed: 12/13/2022] Open
Abstract
Little is known about the role of cell-cell adhesion in the development of mineralized tissues. Here we report that PERP, a tetraspan membrane protein essential for epithelial integrity, regulates enamel formation. PERP is necessary for proper cell attachment and gene expression during tooth development, and its expression is controlled by P63, a master regulator of stratified epithelial development. During enamel formation, PERP is localized to the interface between the enamel-producing ameloblasts and the stratum intermedium (SI), a layer of cells subjacent to the ameloblasts. Perp-null mice display dramatic enamel defects, which are caused, in part, by the detachment of ameloblasts from the SI. Microarray analysis comparing gene expression in teeth of wild-type and Perp-null mice identified several differentially expressed genes during enamel formation. Analysis of these genes in ameloblast-derived LS8 cells upon knockdown of PERP confirmed the role for PERP in the regulation of gene expression. Together, our data show that PERP is necessary for the integrity of the ameloblast-SI interface and that a lack of Perp causes downregulation of genes that are required for proper enamel formation.
Collapse
Affiliation(s)
- Andrew H. Jheon
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California, San Francisco, CA 94143, USA
| | - Pasha Mostowfi
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California, San Francisco, CA 94143, USA
| | - Malcolm L. Snead
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street – CSA 103, Los Angeles, CA 90033, USA
| | - Rebecca A. Ihrie
- Department of Neurological Surgery and Institute for Regeneration Medicine, University of California, San Francisco, 35 Medical Center Way, RMB933, San Francisco, CA 94143-0525, USA
| | - Eli Sone
- Faculty of Dentistry, Department of Materials Science and Engineering, and Institute of Biomaterials and Biomedical Engineering, University of Toronto, M5S 3G9, Canada
| | - Tiziano Pramparo
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| | - Laura D. Attardi
- Departments of Radiation Oncology and Genetics, Stanford University, Palo Alto, CA 94305-5101, USA
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California, San Francisco, CA 94143, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
35
|
Runkle EA, Antonetti DA. The blood-retinal barrier: structure and functional significance. Methods Mol Biol 2011; 686:133-48. [PMID: 21082369 DOI: 10.1007/978-1-60761-938-3_5] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Formation and maintenance of the blood-retinal barrier is required for proper vision and loss of this barrier contributes to the pathology of a wide number of retinal diseases. The retina is responsible for converting visible light into the electrochemical signal interpreted by the brain as vision. Multiple cell types are required for this function, which are organized into eight distinct cell layers. These neural and glial cells gain metabolic support from a unique vascular structure that provides the necessary nutrients while minimizing interference with light sensing. In addition to the vascular contribution, the retina also possesses an epithelial barrier, the retinal pigment epithelium, which is located at the posterior of the eye and controls exchange of nutrients with the choroidal vessels. Together the vascular and epithelial components of the blood-retinal barrier maintain the specialized environment of the neural retina. Both the vascular endothelium and pigment epithelium possess a well-developed junctional complex that includes both adherens and tight junctions conferring a high degree of control of solute and fluid permeability. Understanding induction and regulation of the blood-retinal barrier will allow the development of therapies aimed at restoring the barrier when compromised in disease or allowing the specific transport of therapies across this barrier when needed. This chapter will highlight the anatomical structure of the blood-retinal barrier and explore the molecular structure of the tight junctions that provide the unique barrier properties of the blood--retinal barrier.
Collapse
Affiliation(s)
- E Aaron Runkle
- Department of Ophthalmology, Penn State College of Medicine, Hershey, PA, USA
| | | |
Collapse
|
36
|
Yoshida T, Miyoshi J, Takai Y, Thesleff I. Cooperation of nectin-1 and nectin-3 is required for normal ameloblast function and crown shape development in mouse teeth. Dev Dyn 2010; 239:2558-69. [PMID: 21038445 DOI: 10.1002/dvdy.22395] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Nectins are immunoglobulin-like cell adhesion proteins and their interactions recruit various cell-cell junctions. Mutations in human NECTIN-1 cause an ectodermal dysplasia syndrome, but Nectin-1 null mice have only slight defects in teeth, suggesting compensation by other nectin(s). We observed overlapping expression of nectin-3 with nectin-1 and enamel abnormality in the nectin-3 mutant. We, therefore, generated nectin-1;nectin-3 compound mutants. However, all teeth developed and no significant dental abnormalities were observed before birth. At postnatal day 10, the upper molars of compound mutants exhibited conical crown shape and retarded enamel maturation. Nectin-1 was expressed in ameloblasts whereas nectin-3 was expressed in neighboring stratum intermedium cells at this stage. The immunohistochemical localization and electron microscopical observations indicated that the desmosomal junctions between stratum intermedium and ameloblasts were significantly reduced. These results suggest that heterophilic interaction between nectin-1 and nectin-3 recruits desmosomal junctions, and that these are required for proper enamel formation.
Collapse
|
37
|
Chen X, Koh E, Yoder M, Gumbiner BM. A protocadherin-cadherin-FLRT3 complex controls cell adhesion and morphogenesis. PLoS One 2009; 4:e8411. [PMID: 20027292 PMCID: PMC2791867 DOI: 10.1371/journal.pone.0008411] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 11/30/2009] [Indexed: 11/19/2022] Open
Abstract
Background Paraxial protocadherin (PAPC) and fibronectin leucine-rich domain transmembrane protein-3 (FLRT3) are induced by TGFβ signaling in Xenopus embryos and both regulate morphogenesis by inhibiting C-cadherin mediated cell adhesion. Principal Findings We have investigated the functional and physical relationships between PAPC, FLRT3, and C-cadherin. Although neither PAPC nor FLRT3 are required for each other to regulate C-cadherin adhesion, they do interact functionally and physically, and they form a complex with cadherins. By itself PAPC reduces cell adhesion physiologically to induce cell sorting, while FLRT3 disrupts adhesion excessively to cause cell dissociation. However, when expressed together PAPC limits the cell dissociating and tissue disrupting activity of FLRT3 to make it effective in physiological cell sorting. PAPC counteracts FLRT3 function by inhibiting the recruitment of the GTPase RND1 to the FLRT3 cytoplasmic domain. Conclusions/Significance PAPC and FLRT3 form a functional complex with cadherins and PAPC functions as a molecular “governor” to maintain FLRT3 activity at the optimal level for physiological regulation of C-cadherin adhesion, cell sorting, and morphogenesis.
Collapse
Affiliation(s)
- Xuejun Chen
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, Virginia, United States of America
| | - Eunjin Koh
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, Virginia, United States of America
| | - Michael Yoder
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, Virginia, United States of America
| | - Barry M. Gumbiner
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
38
|
Ooshio T, Kobayashi R, Ikeda W, Miyata M, Fukumoto Y, Matsuzawa N, Ogita H, Takai Y. Involvement of the interaction of afadin with ZO-1 in the formation of tight junctions in Madin-Darby canine kidney cells. J Biol Chem 2009; 285:5003-12. [PMID: 20008323 DOI: 10.1074/jbc.m109.043760] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Tight junctions (TJs) and adherens junctions (AJs) are major junctional apparatuses in epithelial cells. Claudins and junctional adhesion molecules (JAMs) are major cell adhesion molecules (CAMs) at TJs, whereas cadherins and nectins are major CAMs at AJs. Claudins and JAMs are associated with ZO proteins, whereas cadherins are associated with beta- and alpha-catenins, and nectins are associated with afadin. We previously showed that nectins first form cell-cell adhesions where the cadherin-catenin complex is recruited to form AJs, followed by the recruitment of the JAM-ZO and claudin-ZO complexes to the apical side of AJs to form TJs. It is not fully understood how TJ components are recruited to the apical side of AJs. We studied the roles of afadin and ZO-1 in the formation of TJs in Madin-Darby canine kidney (MDCK) cells. Before the formation of TJs, ZO-1 interacted with afadin through the two proline-rich regions of afadin and the SH3 domain of ZO-1. During and after the formation of TJs, ZO-1 dissociated from afadin and associated with JAM-A. Knockdown of afadin impaired the formation of both AJs and TJs in MDCK cells, whereas knockdown of ZO-1 impaired the formation of TJs, but not AJs. Re-expression of full-length afadin restored the formation of both AJs and TJs in afadin-knockdown MDCK cells, whereas re-expression of afadin-DeltaPR1-2, which is incapable of binding to ZO-1, restored the formation of AJs, but not TJs. These results indicate that the transient interaction of afadin with ZO-1 is necessary for the formation of TJs in MDCK cells.
Collapse
Affiliation(s)
- Takako Ooshio
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bielek H, Anselmo A, Dermardirossian C. Morphological and proliferative abnormalities in renal mesangial cells lacking RhoGDI. Cell Signal 2009; 21:1974-83. [PMID: 19765647 DOI: 10.1016/j.cellsig.2009.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 09/09/2009] [Indexed: 11/28/2022]
Abstract
The regulation of Rho GTPase activities and expression is critical in the development and function of the kidney. Rho GTPase activities and cytosol-membrane cycling are regulated by Rho GDP Dissociation Inhibitor (RhoGDI), and RhoGDI knockout mice develop defects in kidney structure and function that lead to death due to renal failure. It is therefore important to understand the changes in RhoGDI-regulated Rho GTPase activities and cell morphology that lead to kidney failure in RhoGDI (-/-) mice. Here, we characterize a renal mesangial cell line derived from the RhoGDI (-/-) mouse in which we verify the absence of GDI proteins. In the absence of RhoGDI, we show an increase in the specific activity of Rac1, and to a lesser extent, RhoA and Cdc42 GTPases in these cells. This is accompanied by a compensatory decrease in the steady-state protein levels of Rho GTPases. Morphological analysis of RhoGDI (-/-) mesangial cells reveals a decrease in cell spreading and in focal contacts compared to wild-type cells. Finally, RhoGDI (-/-) mesangial cells show a decreased ability to proliferate and survive. These functional and structural changes are likely to contribute to the defects in renal architecture and function observed in the RhoGDI (-/-) mouse.
Collapse
Affiliation(s)
- Heike Bielek
- Department of Immunology and Microbial Science, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, United States
| | | | | |
Collapse
|
40
|
Ryu JK, Zhang LW, Jin HR, Piao S, Choi MJ, Tuvshintur B, Tumurbaatar M, Shin SH, Han JY, Kim WJ, Suh JK. Derangements in Endothelial Cell-to-Cell Junctions Involved in the Pathogenesis of Hypercholesterolemia-Induced Erectile Dysfunction. J Sex Med 2009; 6:1893-907. [DOI: 10.1111/j.1743-6109.2009.01275.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
41
|
Severson EA, Lee WY, Capaldo CT, Nusrat A, Parkos CA. Junctional adhesion molecule A interacts with Afadin and PDZ-GEF2 to activate Rap1A, regulate beta1 integrin levels, and enhance cell migration. Mol Biol Cell 2009; 20:1916-25. [PMID: 19176753 DOI: 10.1091/mbc.e08-10-1014] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Junctional adhesion molecule-A (JAM-A) is a transmembrane tight junction protein that has been shown to regulate barrier function and cell migration through incompletely understood mechanisms. We have previously demonstrated that JAM-A regulates cell migration by dimerization of the membrane-distal immunoglobulin-like loop and a C-terminal postsynaptic density 95/disc-large/zona occludens (PDZ) binding motif. Disruption of dimerization resulted in decreased epithelial cell migration secondary to diminished levels of beta1 integrin and active Rap1. Here, we report that JAM-A is physically and functionally associated with the PDZ domain-containing molecules Afadin and PDZ-guanine nucleotide exchange factor (GEF) 2, but not zonula occludens (ZO)-1, in epithelial cells, and these interactions mediate outside-in signaling events. Both Afadin and PDZ-GEF2 colocalized and coimmunoprecipitated with JAM-A. Furthermore, association of PDZ-GEF2 with Afadin was dependent on the expression of JAM-A. Loss of JAM-A, Afadin, or PDZ-GEF2, but not ZO-1 or PDZ-GEF1, similarly decreased cellular levels of activated Rap1, beta1 integrin protein, and epithelial cell migration. The functional effects observed were secondary to decreased levels of Rap1A because knockdown of Rap1A, but not Rap1B, resulted in decreased beta1 integrin levels and reduced cell migration. These findings suggest that JAM-A dimerization facilitates formation of a complex with Afadin and PDZ-GEF2 that activates Rap1A, which regulates beta1 integrin levels and cell migration.
Collapse
Affiliation(s)
- Eric A Severson
- Epithelial Pathobiology Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
42
|
Koshiba H, Hosokawa K, Kubo A, Tokumitsu N, Watanabe A, Honjo H. Junctional Adhesion Molecule: An Expression in Human Endometrial Carcinoma. Int J Gynecol Cancer 2009; 19:208-13. [DOI: 10.1111/igc.0b013e31819bc6e9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Junctional adhesion molecule A (JAM-A) is involved in cell-cell contact and tight junction formation. Loss of cell adhesion molecules may be associated with high histologic grade and invasiveness of endometrial carcinoma. We attempted to determine JAM-A expression in human endometrial carcinoma and its correlations with pathologic features, stage, and survival. Junctional adhesion molecule A expression in human endometrial carcinoma was evaluated by immunohistochemistry. In addition, we cultured human well and poorly differentiated endometrial adenocarcinoma cell lines, Ishikawa cells, and KLE in 3-dimensional basement membrane preparation, and JAM-A expression in these cells was assessed by real-time reverse transcription-polymerase chain reaction and immunohistochemistry. Junctional adhesion molecule A immunostaining intensity was negatively correlated with histologic grade (τ = −0.420, P < 0.0001), myometrial invasion (τ = −0.306, P < 0.01), and stage (τ = −0.383, P < 0.0001). Low JAM-A immunostaining intensity was associated with positive vascular space involvement (P < 0.01). Moreover, low immunostain intensity was significantly (P < 0.0001) related to low overall survival rate and progression-free survival rate. Additionally, in our 3-dimensional epithelial cell culture, JAM-A expression in poorly differentiated adenocarcinoma was significantly lower than that in well-differentiated adenocarcinoma (P < 0.001). Junctional adhesion molecule A expression seems to be reduced in high-grade or advanced endometrial carcinoma and may be a prognostic factor.
Collapse
|
43
|
Brakeman PR, Liu KD, Shimizu K, Takai Y, Mostov KE. Nectin proteins are expressed at early stages of nephrogenesis and play a role in renal epithelial cell morphogenesis. Am J Physiol Renal Physiol 2008; 296:F564-74. [PMID: 19116242 DOI: 10.1152/ajprenal.90328.2008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Development of the nephron requires conversion of the metanephric mesenchyme into tubular epithelial structures with specifically organized intercellular junctions. The nectin proteins are a family of transmembrane proteins that dimerize to form intercellular junctional complexes between epithelial cells. In this study, we demonstrate that nectin junctions appear during the earliest stages of epithelial cell morphogenesis in the murine nephron concurrently with the transition of mesenchymal cells into epithelial cells. We have defined the role of nectin during epithelial cell morphogenesis by studying nectin in a three-dimensional culture of Madin-Darby canine kidney (MDCK) cells. In a three-dimensional culture of MDCK cells grown in purified type 1 collagen, expression of a dominant negative form of nectin causes disruption of the formation of cell polarity and disruption of tight junction (TJ) formation, as measured by zonula occludens-1 (ZO-1) localization. In MDCK cells cultured in Matrigel, exogenous expression of nectin-1 causes disruption of normal epithelial cell cyst formation and decreased apoptosis. These data demonstrate that nectins play an important role in normal epithelial cell morphogenesis and may play a role in mesenchymal-to-epithelial transition during nephrogenesis by providing an antiapoptotic signal and promoting the formation of TJs and cell polarity.
Collapse
Affiliation(s)
- Paul R Brakeman
- Division of Pediatric Nephrology, Department of Pediatrics, University of California, Box 0748, San Francisco, California 94143-0748, USA.
| | | | | | | | | |
Collapse
|
44
|
Takai Y, Ikeda W, Ogita H, Rikitake Y. The immunoglobulin-like cell adhesion molecule nectin and its associated protein afadin. Annu Rev Cell Dev Biol 2008; 24:309-42. [PMID: 18593353 DOI: 10.1146/annurev.cellbio.24.110707.175339] [Citation(s) in RCA: 295] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nectins are immunoglobulin-like cell adhesion molecules (CAMs) that compose a family of four members. Nectins homophilically and heterophilically interact in trans with each other to form cell-cell adhesions. In addition, they heterophilically interact in trans with other immunoglobulin-like CAMs. Nectins bind afadin, an actin filament (F-actin)-binding protein, at its cytoplasmic tail and associate with the actin cytoskeleton. Afadin additionally serves as an adaptor protein by further binding many scaffolding proteins and F-actin-binding proteins and contributes to the association of nectins with other cell-cell adhesion and intracellular signaling systems. Nectins and afadin play roles in the formation of a variety of cell-cell junctions cooperatively with, or independently of, cadherins. Cooperation between nectins and cadherins is required for the formation of cell-cell junctions; cadherins alone are not sufficient. Additionally, nectins regulate many other cellular activities (such as movement, proliferation, survival, differentiation, polarization, and the entry of viruses) in cooperation with other CAMs and cell surface membrane receptors.
Collapse
Affiliation(s)
- Yoshimi Takai
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | | | | | | |
Collapse
|
45
|
Ogita H, Ikeda W, Takai Y. Roles of cell adhesion molecules nectin and nectin-like molecule-5 in the regulation of cell movement and proliferation. J Microsc 2008; 231:455-65. [PMID: 18755001 DOI: 10.1111/j.1365-2818.2008.02058.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In response to chemoattractants, migrating cells form protrusions, such as lamellipodia and filopodia, and structures, such as ruffles over lamellipodia, focal complexes and focal adhesions at leading edges. The formation of these leading edge structures is essential for directional cell movement. Nectin-like molecule-5 (Necl-5) interacts in cis with PDGF receptor and integrin alpha(v)beta(3), and enhances the activation of signalling molecules associated with these transmembrane proteins, which results in the formation of leading edge structures and enhancement of directional cell movement. When migrating cells come into contact with each other, cell-cell adhesion is initiated, resulting in reduced cell velocity. Necl-5 first interacts in trans with nectin-3. This interaction is transient and induces down-regulation of Necl-5 expression at the cell surface, resulting in reduced cell movement. Cell proliferation is also suppressed by the down-regulation of Necl-5, because the inhibitory effect of Necl-5 on Sprouty2, a negative regulator of the Ras signalling, is diminished. PDGF receptor and integrin alpha(v)beta(3), which have interacted with Necl-5, then form a complex with nectin, which initiates cell-cell adhesion and recruits cadherin to the nectin-based cell-cell adhesion sites to form stable adherens junctions. The formation of adherens junctions stops cell movement, in part through inactivation of integrin alpha(v)beta(3) caused by the trans-interaction of nectin. Thus, nectin and Necl-5 play key roles in the regulation of cell movement and proliferation.
Collapse
Affiliation(s)
- H Ogita
- Department of Molecular Biology and Biochemistry, Faculty of MedicineOsaka, University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | |
Collapse
|
46
|
Barron MJ, Brookes SJ, Draper CE, Garrod D, Kirkham J, Shore RC, Dixon MJ. The cell adhesion molecule nectin-1 is critical for normal enamel formation in mice. Hum Mol Genet 2008; 17:3509-20. [PMID: 18703497 PMCID: PMC2572697 DOI: 10.1093/hmg/ddn243] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2008] [Accepted: 08/12/2008] [Indexed: 12/19/2022] Open
Abstract
Nectin-1 is a member of a sub-family of immunoglobulin-like adhesion molecules and a component of adherens junctions. In the current study, we have shown that mice lacking nectin-1 exhibit defective enamel formation in their incisor teeth. Although the incisors of nectin-1-null mice were hypomineralized, the protein composition of the enamel matrix was unaltered. While strong immunostaining for nectin-1 was observed at the interface between the maturation-stage ameloblasts and the underlying cells of the stratum intermedium (SI), its absence in nectin-1-null mice correlated with separation of the cell layers at this interface. Numerous, large desmosomes were present at this interface in wild-type mice; however, where adhesion persisted in the mutant mice, the desmosomes were smaller and less numerous. Nectins have been shown to regulate tight junction formation; however, this is the first report showing that they may also participate in the regulation of desmosome assembly. Importantly, our results show that integrity of the SI-ameloblast interface is essential for normal enamel mineralization.
Collapse
Affiliation(s)
- Martin J. Barron
- Faculty of Life Sciences
- Dental School, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Steven J. Brookes
- Department of Oral Biology, Leeds Dental Institute, University of Leeds, Clarendon Way, Leeds LS2 9LU, UK
| | | | | | - Jennifer Kirkham
- Department of Oral Biology, Leeds Dental Institute, University of Leeds, Clarendon Way, Leeds LS2 9LU, UK
| | - Roger C. Shore
- Department of Oral Biology, Leeds Dental Institute, University of Leeds, Clarendon Way, Leeds LS2 9LU, UK
| | - Michael J. Dixon
- Faculty of Life Sciences
- Dental School, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
47
|
Kuramitsu K, Ikeda W, Inoue N, Tamaru Y, Takai Y. Novel role of nectin: implication in the co-localization of JAM-A and claudin-1 at the same cell-cell adhesion membrane domain. Genes Cells 2008; 13:797-805. [PMID: 18547333 DOI: 10.1111/j.1365-2443.2008.01206.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tight junctions (TJs) are formed at the apical side of adherens junctions (AJs) in epithelial cells. Major cell adhesion molecules (CAMs) at TJs are JAM and claudin, whereas major CAMs at AJs are nectin and cadherin. We previously showed that nectin initially forms cell-cell adhesion and then recruits cadherin to the nectin-based cell-cell adhesion sites to form AJs, followed by the recruitment of JAM and claudin to the apical side of AJs to form TJs. We investigated the roles of nectin in the formation of TJs by expressing various combinations of CAMs in L fibroblasts with no TJs or AJs. Co-expression of one of the AJ CAMs and one of the TJ CAMs formed two separate cell-cell adhesion membrane domains (CAMDs). Co-expression of nectin-3 and E-cadherin formed the same CAMD, but co-expression of JAM-A and claudin-1 did not form the same CAMD. Co-expression of JAM-A and claudin-1 with nectin-3, but not E-cadherin, made them form the same CAMD, which was separated from the nectin-based CAMD. Nectin-3 required afadin, a nectin- and F-actin-binding protein, for this ability. In conclusion, nectin plays a novel role in the co-localization of JAM and claudin at the same CAMD.
Collapse
Affiliation(s)
- Kaori Kuramitsu
- Department of Molecular Biology and Biochemistry, Osaka Graduate School of Medicine/Faculty of Medicine, Suita 565-0871, Osaka, Japan
| | | | | | | | | |
Collapse
|
48
|
Mruk DD, Silvestrini B, Cheng CY. Anchoring junctions as drug targets: role in contraceptive development. Pharmacol Rev 2008; 60:146-80. [PMID: 18483144 PMCID: PMC3023124 DOI: 10.1124/pr.107.07105] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In multicellular organisms, cell-cell interactions are mediated in part by cell junctions, which underlie tissue architecture. Throughout spermatogenesis, for instance, preleptotene leptotene spermatocytes residing in the basal compartment of the seminiferous epithelium must traverse the blood-testis barrier to enter the adluminal compartment for continued development. At the same time, germ cells must also remain attached to Sertoli cells, and numerous studies have reported extensive restructuring at the Sertoli-Sertoli and Sertoli-germ cell interface during germ cell movement across the seminiferous epithelium. Furthermore, the proteins and signaling cascades that regulate adhesion between testicular cells have been largely delineated. These findings have unveiled a number of potential "druggable" targets that can be used to induce premature release of germ cells from the seminiferous epithelium, resulting in transient infertility. Herein, we discuss a novel approach with the aim of developing a nonhormonal male contraceptive for future human use, one that involves perturbing adhesion between Sertoli and germ cells in the testis.
Collapse
Affiliation(s)
- Dolores D Mruk
- Population Council, Center for Biomedical Research, The Mary M Wohlford Laboratory for Male Contraceptive Research, 1230 York Avenue, New York, NY 10065, USA.
| | | | | |
Collapse
|
49
|
Ogita H, Takai Y. Cross-talk among integrin, cadherin, and growth factor receptor: roles of nectin and nectin-like molecule. ACTA ACUST UNITED AC 2008; 265:1-54. [PMID: 18275885 DOI: 10.1016/s0074-7696(07)65001-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Integrin, cadherin, and growth factor receptor are key molecules for fundamental cellular functions including cell movement, proliferation, differentiation, adhesion, and survival. These cell surface molecules cross-talk with each other in the regulation of such cellular functions. Nectin and nectin-like molecule (Necl) have been identified as cell adhesion molecules that belong to the immunoglobulin superfamily. Nectin and Necl play important roles in the integration of integrin, cadherin, and growth factor receptor at the cell-cell adhesion sites of contacting cells and at the leading edges of moving cells, and thus are also involved in the fundamental cellular functions together with integrin, cadherin, and growth factor receptor. This chapter describes how newly identified cell adhesion molecules, nectin and Necl, modulate the cross-talk among integrin, cadherin, and growth factor receptor and how these integrated molecules act in the regulation of fundamental cellular functions.
Collapse
Affiliation(s)
- Hisakazu Ogita
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine/Faculty of Medicine, Suita, Osaka 565-0871, Japan
| | | |
Collapse
|
50
|
Lim ST, Lim KC, Giuliano RE, Federoff HJ. Temporal and spatial localization of nectin-1 and l-afadin during synaptogenesis in hippocampal neurons. J Comp Neurol 2008; 507:1228-44. [PMID: 18181141 DOI: 10.1002/cne.21608] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Nectins are cell adhesion molecules that, together with the intracellular binding partner afadin, mediate adhesion and signaling at a variety of intercellular junctions. In this work we studied the distribution of nectin-1 and afadin during hippocampal synapse formation using cultured primary hippocampal neurons. Nectin-1 and afadin cluster at developing synapses between hippocampal neurons. These nectin-afadin clusters uniformly colocalize with N-cadherin-catenin pairs, suggesting that formation of developing synapses involves participation of both bimolecular systems. Nectin-1 is initially expressed at excitatory and inhibitory synapses but is progressively lost at inhibitory synapses during their maturation. Treatment of neurons with actin depolymerizing agents disrupts the synaptically localized nectin-1 and afadin cluster at an early stage and elicits nectin-1 ectodomain shedding. These data indicate that the synaptic localization of nectin-1 and l-afadin are F-actin-dependent and that the shedding of nectin-1 is a mechanism contributing to synaptic plasticity.
Collapse
Affiliation(s)
- Seung T Lim
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | | |
Collapse
|