151
|
Murakawa H, Togashi H. Continuous models for cell–cell adhesion. J Theor Biol 2015; 374:1-12. [DOI: 10.1016/j.jtbi.2015.03.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 02/14/2015] [Accepted: 03/03/2015] [Indexed: 11/24/2022]
|
152
|
Yamamoto T, Mori T, Sawada M, Matsushima H, Ito F, Akiyama M, Kitawaki J. Loss of AF-6/afadin induces cell invasion, suppresses the formation of glandular structures and might be a predictive marker of resistance to chemotherapy in endometrial cancer. BMC Cancer 2015; 15:275. [PMID: 25879875 PMCID: PMC4399104 DOI: 10.1186/s12885-015-1286-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 03/30/2015] [Indexed: 02/04/2023] Open
Abstract
Background AF-6/afadin plays an important role in the formation of adherence junctions. In breast and colon cancer, loss of AF-6/afadin induces cell migration and cell invasion. We aimed to elucidate the role of AF-6/afadin in human endometrial cancer. Methods Morphology and AF-6/afadin expression in endometrial cancer cell lines was investigated by 3-dimensional culture. We used Matrigel invasion assay to demonstrate AF-6/afadin knockdown induced invasive capability. Cell proliferation assay was performed to estimate chemoresistance to doxorubicin, paclitaxel and cisplatin induced by AF-6/afadin knockdown. The associations between AF-6/afadin expression and clinicopathological status were determined by immunohistochemical analysis in endometrial cancer tissues. Informed consent was obtained from all patients before the study. Results The majority of cell clumps in 3-dimensional cultures of Ishikawa cells that strongly expressed AF-6/afadin showed round gland-like structures. In contrast, the cell clumps in 3-dimensional cultures of HEC1A and AN3CA cells—both weakly expressing AF-6/afadin—showed irregular gland-like structures and disorganized colonies with no gland-like structures, respectively. AF-6/afadin knockdown resulted in reduced number of gland-like structures in 3-dimensional cultures and enhancement of cell invasion and phosphorylation of ERK1/2 and Src in the highly AF-6/afadin-expressing endometrial cancer cell line. Inhibitors of MAPK/ERK kinase (MEK) (U0126) and Src (SU6656) suppressed the AF-6/afadin knockdown-induced invasive capability. AF-6/afadin knockdown induced chemoresistance to doxorubicin, paclitaxel and cisplatin in Ishikawa cells, not in HEC1A. Immunohistochemical analysis showed that AF-6/afadin expression was significantly associated with myometrial invasion and high histological grade. Conclusions AF-6/afadin regulates cell morphology and invasiveness. Invasive capability is partly regulated through the ERK and Src pathway. The inhibitors to these pathways might be molecular-targeted drugs which suppress myometrial invasion in endometrial cancer. AF-6/afadin could be a useful selection marker for fertility-sparing therapy for patients with atypical hyperplasia or grade 1 endometrioid adenocarcinoma with no myometrial invasion. AF-6/afadin knockdown induced chemoresistance especially to cisplatin. Therefore, loss of AF-6/afadin might be a predictive marker of chemoresistance to cisplatin.
Collapse
Affiliation(s)
- Takuro Yamamoto
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Taisuke Mori
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Morio Sawada
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Hiroshi Matsushima
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Fumitake Ito
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Makoto Akiyama
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Jo Kitawaki
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
153
|
Actin-tethered junctional complexes in angiogenesis and lymphangiogenesis in association with vascular endothelial growth factor. BIOMED RESEARCH INTERNATIONAL 2015; 2015:314178. [PMID: 25883953 PMCID: PMC4389985 DOI: 10.1155/2015/314178] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/23/2014] [Accepted: 10/31/2014] [Indexed: 12/21/2022]
Abstract
Vasculature is present in all tissues and therefore is indispensable for development, biology, and pathology of multicellular organisms. Endothelial cells guarantee proper function of the vessels and are the original component in angiogenesis. Morphogenesis of the vascular system utilizes processes like cell adhesion, motility, proliferation, and survival that are closely related to the dynamics of actin filaments and actin-tethered adhesion complexes. Here we review involvement of actin cytoskeleton-associated junctional molecules of endothelial cells in angiogenesis and lymphangiogenesis. Particularly, we focus on F-actin binding protein afadin, an adaptor protein involved in broad range of signaling mechanisms. Afadin mediates the pathways of vascular endothelial growth factor- (VEGF-) and sphingosine 1-phosphate-triggered angiogenesis and is essential for embryonic development of lymph vessels in mice. We propose that targeting actin-tethered junctional molecules, including afadin, may present a new approach to angiogenic therapy that in combination with today used medications like VEGF inhibitors will benefit against development of pathological angiogenesis.
Collapse
|
154
|
Proteomic analysis of proteins surrounding occludin and claudin-4 reveals their proximity to signaling and trafficking networks. PLoS One 2015; 10:e0117074. [PMID: 25789658 PMCID: PMC4366163 DOI: 10.1371/journal.pone.0117074] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/18/2014] [Indexed: 01/15/2023] Open
Abstract
Tight junctions are complex membrane structures that regulate paracellular movement of material across epithelia and play a role in cell polarity, signaling and cytoskeletal organization. In order to expand knowledge of the tight junction proteome, we used biotin ligase (BioID) fused to occludin and claudin-4 to biotinylate their proximal proteins in cultured MDCK II epithelial cells. We then purified the biotinylated proteins on streptavidin resin and identified them by mass spectrometry. Proteins were ranked by relative abundance of recovery by mass spectrometry, placed in functional categories, and compared not only among the N- and C- termini of occludin and the N-terminus of claudin-4, but also with our published inventory of proteins proximal to the adherens junction protein E-cadherin and the tight junction protein ZO-1. When proteomic results were analyzed, the relative distribution among functional categories was similar between occludin and claudin-4 proximal proteins. Apart from already known tight junction- proteins, occludin and claudin-4 proximal proteins were enriched in signaling and trafficking proteins, especially endocytic trafficking proteins. However there were significant differences in the specific proteins comprising the functional categories near each of the tagging proteins, revealing spatial compartmentalization within the junction complex. Taken together, these results expand the inventory of known and unknown proteins at the tight junction to inform future studies of the organization and physiology of this complex structure.
Collapse
|
155
|
Raza SI, Nasser Dar R, Shah AA, Ahmad W. A novel homozygous nonsense mutation in the PVRL4 gene and expansion of clinical spectrum of EDSS1. Ann Hum Genet 2015; 79:92-8. [PMID: 25529316 DOI: 10.1111/ahg.12094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/17/2014] [Indexed: 12/01/2022]
Abstract
Ectodermal dysplasias (EDs) belong to a group of genetic diseases which result from alterations in ectoderm-derived appendages including hair, nail, teeth and sweat glands. Ectodermal dysplasia syndactyly syndrome (EDSS1) is one of the rare forms of ED caused by mutations in nectin-4, encoded by the PVRL4 gene. The present study described clinical investigation of the EDSS1 identified in a large consanguineous family. Furthermore, DNA sequence analysis revealed a novel homozygous nonsense mutation (181C>T, p.Asp61*) in the PVRL4 gene.
Collapse
Affiliation(s)
- Syed Irfan Raza
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; Army Medical College, National University of Science & Technology (NUST), Islamabad, Pakistan
| | | | | | | |
Collapse
|
156
|
Gehren AS, Rocha MR, de Souza WF, Morgado-Díaz JA. Alterations of the apical junctional complex and actin cytoskeleton and their role in colorectal cancer progression. Tissue Barriers 2015; 3:e1017688. [PMID: 26451338 DOI: 10.1080/21688370.2015.1017688] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/31/2015] [Accepted: 02/06/2015] [Indexed: 01/08/2023] Open
Abstract
Colorectal cancer represents the fourth highest mortality rate among cancer types worldwide. An understanding of the molecular mechanisms that regulate their progression can prevents or reduces mortality due to this disease. Epithelial cells present an apical junctional complex connected to the actin cytoskeleton, which maintains the dynamic properties of this complex, tissue architecture and cell homeostasis. Several studies have indicated that apical junctional complex alterations and actin cytoskeleton disorganization play a critical role in epithelial cancer progression. However, few studies have examined the existence of an interrelation between these 2 components, particularly in colorectal cancer. This review discusses the recent progress toward elucidating the role of alterations of apical junctional complex constituents and of modifications of actin cytoskeleton organization and discusses how these events are interlinked to modulate cellular responses related to colorectal cancer progression toward successful metastasis.
Collapse
Affiliation(s)
- Adriana Sartorio Gehren
- Program of Cellular Biology; Brazilian National Cancer Institute (INCA) ; Rio de Janeiro, Brazil
| | - Murilo Ramos Rocha
- Program of Cellular Biology; Brazilian National Cancer Institute (INCA) ; Rio de Janeiro, Brazil
| | | | - José Andrés Morgado-Díaz
- Program of Cellular Biology; Brazilian National Cancer Institute (INCA) ; Rio de Janeiro, Brazil
| |
Collapse
|
157
|
Izumi H, Hirabayashi K, Nakamura N, Nakagohri T. Nectin expression in pancreatic adenocarcinoma: nectin-3 is associated with a poor prognosis. Surg Today 2015; 45:487-94. [PMID: 25690753 PMCID: PMC4359290 DOI: 10.1007/s00595-015-1126-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/20/2015] [Indexed: 01/09/2023]
Abstract
Purpose Nectins are cell adhesion molecules that regulate the formation of adherens junctions and are linked with E-cadherin-based cell–cell adherens junctions. In pancreatic cancer, the expression of E-cadherin and nectins is considered to be related to metastasis, invasion and prognosis. Methods We evaluated the distribution of cells that were positive for nectin subtypes and E-cadherin using immunohistochemistry in specimens of human pancreatic adenocarcinoma, and correlated these results with the clinicopathological features and patient outcomes. Results The immunohistochemical distribution of nectin-1 and E-cadherin showed a good correlation (r = 0.523, p < 0.01). Tumors over 4 cm in diameter had more intense staining for nectin-4 than smaller tumors (p = 0.035). Nectin-2 expression correlated with a poorer histological grade (p = 0.04). The cases that showed diffuse nectin-3 expression had a better prognosis than those with negative expression (p = 0.018). Conclusion Our results showed that the expression of nectin-3 in pancreatic cancer can be a prognostic factor.
Collapse
Affiliation(s)
- Hideki Izumi
- Department of Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Kenichi Hirabayashi
- Department of Pathology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193 Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193 Japan
| | - Toshio Nakagohri
- Department of Surgery, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
158
|
Samanta D, Almo SC. Nectin family of cell-adhesion molecules: structural and molecular aspects of function and specificity. Cell Mol Life Sci 2015; 72:645-58. [PMID: 25326769 PMCID: PMC11113404 DOI: 10.1007/s00018-014-1763-4] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/11/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022]
Abstract
Cell-cell adhesive processes are central to the physiology of multicellular organisms. A number of cell surface molecules contribute to cell-cell adhesion, and the dysfunction of adhesive processes underlies numerous developmental defects and inherited diseases. The nectins, a family of four immunoglobulin superfamily members (nectin-1 to -4), interact through their extracellular domains to support cell-cell adhesion. While both homophilic and heterophilic interactions among the nectins are implicated in cell-cell adhesion, cell-based and biochemical studies suggest heterophilic interactions are stronger than homophilic interactions and control a range of physiological processes. In addition to interactions within the nectin family, heterophilic associations with nectin-like molecules, immune receptors, and viral glycoproteins support a wide range of biological functions, including immune modulation, cancer progression, host-pathogen interactions and immune evasion. We review current structural and molecular knowledge of nectin recognition processes, with a focus on the biochemical and biophysical determinants of affinity and selectivity that drive distinct nectin associations. These proteins and interactions are discussed as potential targets for immunotherapy.
Collapse
Affiliation(s)
- Dibyendu Samanta
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| |
Collapse
|
159
|
Birukova AA, Meng F, Tian Y, Meliton A, Sarich N, Quilliam LA, Birukov KG. Prostacyclin post-treatment improves LPS-induced acute lung injury and endothelial barrier recovery via Rap1. Biochim Biophys Acta Mol Basis Dis 2014; 1852:778-91. [PMID: 25545047 DOI: 10.1016/j.bbadis.2014.12.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/15/2014] [Accepted: 12/22/2014] [Indexed: 12/27/2022]
Abstract
Protective effects of prostacyclin (PC) or its stable analog beraprost against agonist-induced lung vascular inflammation have been associated with elevation of intracellular cAMP and Rac GTPase signaling which inhibited the RhoA GTPase-dependent pathway of endothelial barrier dysfunction. This study investigated a distinct mechanism of PC-stimulated lung vascular endothelial (EC) barrier recovery and resolution of LPS-induced inflammation mediated by small GTPase Rap1. Efficient barrier recovery was observed in LPS-challenged pulmonary EC after prostacyclin administration even after 15 h of initial inflammatory insult and was accompanied by the significant attenuation of p38 MAP kinase and NFκB signaling and decreased production of IL-8 and soluble ICAM1. These effects were reproduced in cells post-treated with 8CPT, a small molecule activator of Rap1-specific nucleotide exchange factor Epac. By contrast, pharmacologic Epac inhibitor, Rap1 knockdown, or knockdown of cell junction-associated Rap1 effector afadin attenuated EC recovery caused by PC or 8CPT post-treatment. The key role of Rap1 in lung barrier restoration was further confirmed in the murine model of LPS-induced acute lung injury. Lung injury was monitored by measurements of bronchoalveolar lavage protein content, cell count, and Evans blue extravasation and live imaging of vascular leak over 6 days using a fluorescent tracer. The data showed significant acceleration of lung recovery by PC and 8CPT post-treatment, which was abrogated in Rap1a(-/-) mice. These results suggest that post-treatment with PC triggers the Epac/Rap1/afadin-dependent mechanism of endothelial barrier restoration and downregulation of p38MAPK and NFκB inflammatory cascades, altogether leading to accelerated lung recovery.
Collapse
Affiliation(s)
- Anna A Birukova
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Fanyong Meng
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Yufeng Tian
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Angelo Meliton
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Nicolene Sarich
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Lawrence A Quilliam
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202-5122, USA
| | - Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
160
|
Mateo M, Navaratnarajah CK, Willenbring RC, Maroun JW, Iankov I, Lopez M, Sinn PL, Cattaneo R. Different roles of the three loops forming the adhesive interface of nectin-4 in measles virus binding and cell entry, nectin-4 homodimerization, and heterodimerization with nectin-1. J Virol 2014; 88:14161-71. [PMID: 25275122 PMCID: PMC4249131 DOI: 10.1128/jvi.02379-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Many viruses utilize cell adhesion molecules of the immunoglobulin superfamily as receptors. In particular, viruses of different classes exploit nectins. The large DNA viruses, herpes simplex and pseudorabies viruses, use ubiquitous nectins 1 and 2. The negative-strand RNA virus measles virus (MeV) uses tissue-specific nectin-4, and the positive-strand RNA virus poliovirus uses nectin-like 5 (necl-5), also known as poliovirus receptor. These viruses contact the BC, C'C", and FG loops on the upper tip of their receptor's most membrane-distal domain. This location corresponds to the newly defined canonical adhesive interface of nectins, but how viruses utilize this interface has remained unclear. Here we show that the same key residues in the BC and FG loops of nectin-4 govern binding to the MeV attachment protein hemagglutinin (H) and cell entry, nectin-4 homodimerization, and heterodimerization with nectin-1. On the other hand, residues in the C'C" loop necessary for homo- and heterotypic interactions are dispensable for MeV-induced fusion and cell entry. Remarkably, the C'C" loop governs dissociation of the nectin-4 and H ectodomains. We provide formal proof that H can interfere with the formation of stable nectin-1/nectin-4 heterodimers. Finally, while developing an alternative model to study MeV spread, we observed that polarized primary pig airway epithelial sheets cannot be infected. We show that a single amino acid variant in the BC loop of pig nectin-4 fully accounts for restricted MeV entry. Thus, the three loops forming the adhesive interface of nectin-4 have different roles in supporting MeV H association and dissociation and MeV-induced fusion. IMPORTANCE Different viruses utilize nectins as receptors. Nectins are immunoglobulin superfamily glycoproteins that mediate cell-cell adhesion in vertebrate tissues. They interact through an adhesive interface located at the top of their membrane-distal domain. How viruses utilize the three loops forming this interface has remained unclear. We demonstrate that while nectin-nectin interactions require residues in all three loops, the association of nectin-4 with the measles virus hemagglutinin requires only the BC and FG loops. However, we discovered that residues in the C'C" loop modulate the dissociation of nectin-4 from the viral hemagglutinin. Analogous mechanisms may support cell entry of other viruses that utilize nectins or other cell adhesion molecules of the immunoglobulin superfamily as receptors.
Collapse
Affiliation(s)
- Mathieu Mateo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Robin C Willenbring
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA Virology and Gene Therapy track, Mayo Graduate School, Rochester, Minnesota, USA
| | - Justin W Maroun
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA Virology and Gene Therapy track, Mayo Graduate School, Rochester, Minnesota, USA
| | - Ianko Iankov
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Marc Lopez
- INSERM, UMR1068/CRCM, Institut Paoli-Calmettes and University of Aix-Marseille, Marseille, France
| | - Patrick L Sinn
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA Virology and Gene Therapy track, Mayo Graduate School, Rochester, Minnesota, USA
| |
Collapse
|
161
|
Fujita A, Inanobe A, Hibino H, Nielsen S, Ottersen OP, Kurachi Y. Clustering of Kir4.1 at specialized compartments of the lateral membrane in ependymal cells of rat brain. Cell Tissue Res 2014; 359:627-634. [PMID: 25380566 DOI: 10.1007/s00441-014-2030-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/09/2014] [Indexed: 10/24/2022]
Abstract
Brain ependymal cells, which form an epithelial layer covering the cerebral ventricles, have been shown to play a role in the regulation of cerebrospinal and interstitial fluids. The machinery underlying this, however, remains largely unknown. Here, we report the specific localization of an inwardly rectifying K(+) channel, Kir4.1, on the ependymal cell membrane suggesting involvement of the channel in this function. Immunohistochemical study with confocal microscopy identified Kir4.1 labeling on the lateral but not apical membrane of ependymal cells. Ultrastructural analysis revealed that Kir4.1-immunogold particles were specifically localized and clustered on adjacent membranes at puncta adherens type junctions, whereas an aquaporin water channel, AQP4, that was also detected on the lateral membrane only occurred at components other than adherens junctions. Therefore, in ependymal cells, Kir4.1 and AQP4 are partitioned into distinct membrane compartments that might respectively transport either K(+) or water. Kir4.1 was also expressed in a specialized form of ependymal cell, namely the tanycyte, being abundant in tanycyte processes wrapping neuropils and blood vessels. These specific localizations suggest that Kir4.1 mediates intercellular K(+) exchange between ependymal cells and also K(+)-buffering transport via tanycytes that can interconnect neurons and vessels/ventricles. We propose that ependymal cells and tanycytes differentially operate Kir4.1 and AQP4 actively to control the property of fluids at local areas in the brain.
Collapse
Affiliation(s)
- Akikazu Fujita
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan. .,Field of Veterinary Pathobiology, Basic Veterinary Science, Department of Veterinary Medicine, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima, 890-0065, Japan.
| | - Atsushi Inanobe
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Hibino
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.,Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Søren Nielsen
- Water and Salt Research Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ole Petter Ottersen
- Center for Molecular Biology and Neuroscience and Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Yoshihisa Kurachi
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
162
|
Hall JV, Sun J, Slade J, Kintner J, Bambino M, Whittimore J, Schoborg RV. Host nectin-1 is required for efficient Chlamydia trachomatis serovar E development. Front Cell Infect Microbiol 2014; 4:158. [PMID: 25414835 PMCID: PMC4222120 DOI: 10.3389/fcimb.2014.00158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/16/2014] [Indexed: 11/13/2022] Open
Abstract
Interaction of Herpes Simplex Virus (HSV) glycoprotein D (gD) with the host cell surface during Chlamydia trachomatis/HSV co-infection stimulates chlamydiae to become persistent. During viral entry, gD interacts with one of 4 host co-receptors: HVEM (herpes virus entry mediator), nectin-1, nectin-2 and 3-O-sulfated heparan sulfate. HVEM and nectin-1 are high-affinity entry receptors for both HSV-1 and HSV-2. Nectin-2 mediates HSV-2 entry but is inactive for HSV-1, while 3-O-sulfated heparan sulfate facilitates HSV-1, but not HSV-2, entry. Western blot and RT-PCR analyses demonstrate that HeLa and HEC-1B cells express nectin-1 and nectin-2, but not HVEM. Because both HSV-1 and HSV-2 trigger persistence, these data suggest that nectin-1 is the most likely co-receptor involved. Co-infections with nectin-1 specific HSV-1 mutants stimulate chlamydial persistence, as evidenced by aberrant body (AB) formation and decreased production of elementary bodies (EBs). These data indicate that nectin-1 is involved in viral-induced chlamydial persistence. However, inhibition of signal transduction molecules associated with HSV attachment and entry does not rescue EB production during C. trachomatis/HSV-2 co-infection. HSV attachment also does not activate Cdc42 in HeLa cells, as would be expected with viral stimulated activation of nectin-1 signaling. Additionally, immunofluorescence assays confirm that HSV infection decreases nectin-1 expression. Together, these observations suggest that gD binding-induced loss of nectin-1 signaling negatively influences chlamydial growth. Chlamydial infection studies in nectin-1 knockdown (NKD) HeLa cell lines support this hypothesis. In NKD cells, chlamydial inclusions are smaller in size, contain ABs, and produce significantly fewer infectious EBs compared to C. trachomatis infection in control HeLa cells. Overall, the current study indicates that the actions of host molecule, nectin-1, are required for successful C. trachomatis development.
Collapse
Affiliation(s)
- Jennifer V Hall
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA ; Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA
| | - Jingru Sun
- College of Medical Sciences, Washington State University Spokane, WA, USA
| | - Jessica Slade
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA ; Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA
| | - Jennifer Kintner
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA
| | - Marissa Bambino
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA
| | - Judy Whittimore
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA
| | - Robert V Schoborg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA ; Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University Johnson City, TN, USA
| |
Collapse
|
163
|
Ramuz M, Hama A, Huerta M, Rivnay J, Leleux P, Owens RM. Combined optical and electronic sensing of epithelial cells using planar organic transistors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:7083-90. [PMID: 25179835 PMCID: PMC4489338 DOI: 10.1002/adma.201401706] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/28/2014] [Indexed: 05/17/2023]
Abstract
A planar, conducting-polymer-based transistor for combined optical and electronic monitoring of live cells provides a unique platform for monitoring the health of cells in vitro. Monitoring of MDCK-I epithelial cells over several days is shown, along with a demonstration of the device for toxicology studies, of use in future drug discovery or diagnostics applications.
Collapse
Affiliation(s)
- Marc Ramuz
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines CMP-EMSE, MOC880 avenue de Mimet, 13541, Gardanne, France E-mail:
| | - Adel Hama
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines CMP-EMSE, MOC880 avenue de Mimet, 13541, Gardanne, France E-mail:
| | - Miriam Huerta
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines CMP-EMSE, MOC880 avenue de Mimet, 13541, Gardanne, France E-mail:
| | - Jonathan Rivnay
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines CMP-EMSE, MOC880 avenue de Mimet, 13541, Gardanne, France E-mail:
| | - Pierre Leleux
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines CMP-EMSE, MOC880 avenue de Mimet, 13541, Gardanne, France E-mail:
- Aix-Marseille Université, Institut de Neurosciences des Systèmes13005, Marseille, France
- Inserm, UMR_S 110613005, Marseille, France
- Microvitae Technologies, Pôle d’Activité Y. Morandat13120, Gardanne, France
| | - Róisín M Owens
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines CMP-EMSE, MOC880 avenue de Mimet, 13541, Gardanne, France E-mail:
| |
Collapse
|
164
|
Tanaka-Okamoto M, Itoh Y, Miyoshi J, Mizoguchi A, Mizutani K, Takai Y, Inoue M. Genetic ablation of afadin causes mislocalization and deformation of Paneth cells in the mouse small intestinal epithelium. PLoS One 2014; 9:e110549. [PMID: 25333284 PMCID: PMC4204899 DOI: 10.1371/journal.pone.0110549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/15/2014] [Indexed: 12/23/2022] Open
Abstract
Afadin is an actin filament-binding protein that acts cooperatively in cell adhesion with the cell adhesion molecule nectin, and in directional cell movement with the small G protein Rap1 in a nectin-independent manner. We studied the role of afadin in the organization of the small intestinal epithelium using afadin conditional gene knockout (cKO) mice. Afadin was localized at adherens junctions of all types of epithelial cells throughout the crypt-villus axis. Paneth cells were localized at the base of the crypt in control mice, but not confined there, and migrated into the villi in afadin-cKO mice. The distribution of other types of epithelial cells did not change significantly in the mutant mice. The Paneth cells remaining in the crypt exhibited abnormal shapes, were buried between adjacent cells, and did not face the lumen. In these cells, the formation of adherens junctions and tight junctions was impaired. Rap1 and EphB3 were highly expressed in control Paneth cells but markedly down-regulated in the afadin-deficient Paneth cells. Taken together, the results indicate that afadin plays a role in the restricted localization of Paneth cells at the base of the crypt by maintaining their adhesion to adjacent crypt cells and inhibiting their movement toward the top of villi.
Collapse
Affiliation(s)
- Miki Tanaka-Okamoto
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
- * E-mail: (MT-O); (MI)
| | - Yu Itoh
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
- Department of Neural Regeneration and Cell Communication, Mie University Graduate School of Medicine, Mie, Japan
| | - Jun Miyoshi
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
| | - Akira Mizoguchi
- Department of Neural Regeneration and Cell Communication, Mie University Graduate School of Medicine, Mie, Japan
| | - Kiyohito Mizutani
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Disease, Osaka, Japan
- * E-mail: (MT-O); (MI)
| |
Collapse
|
165
|
Defects in the adherens junction complex (E-cadherin/ β-catenin) in inflammatory bowel disease. Cell Tissue Res 2014; 360:749-60. [DOI: 10.1007/s00441-014-1994-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/21/2014] [Indexed: 01/27/2023]
|
166
|
Quiros M, Nusrat A. RhoGTPases, actomyosin signaling and regulation of the epithelial Apical Junctional Complex. Semin Cell Dev Biol 2014; 36:194-203. [PMID: 25223584 DOI: 10.1016/j.semcdb.2014.09.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 08/28/2014] [Accepted: 09/04/2014] [Indexed: 12/22/2022]
Abstract
Epithelial cells form regulated and selective barriers between distinct tissue compartments. The Apical Junctional Complex (AJC) consisting of the tight junction (TJ) and adherens junction (AJ) control epithelial homeostasis, paracellular permeability and barrier properties. The AJC is composed of mutliprotein complexes consisting of transmembrane proteins that affiliate with an underlying perijunctional F-actin myosin ring through cytoplasmic scaffold proteins. AJC protein associations with the apical actin-myosin cytoskeleton are tightly controlled by a number of signaling proteins including the Rho family of GTPases that orchestrate junctional biology, epithelial homeostasis and barrier function. This review highlights the vital relationship of Rho GTPases and AJCs in controlling the epithelial barrier. The pathophysiologic relationship of Rho GTPases, AJC, apical actomyosin cytoskeleton and epithelial barrier function is discussed.
Collapse
Affiliation(s)
- Miguel Quiros
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
167
|
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
|
168
|
Sun TT, Wang Y, Cheng H, Xiao HZ, Xiang JJ, Zhang JT, Yu SBS, Martin TA, Ye L, Tsang LL, Jiang WG, Xiaohua J, Chan HC. Disrupted interaction between CFTR and AF-6/afadin aggravates malignant phenotypes of colon cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:618-28. [PMID: 24373847 DOI: 10.1016/j.bbamcr.2013.12.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 11/26/2013] [Accepted: 12/16/2013] [Indexed: 12/16/2022]
Abstract
How mutations or dysfunction of CFTR may increase the risk of malignancies in various tissues remains an open question. Here we report the interaction between CFTR and an adherens junction molecule, AF-6/afadin, and its involvement in the development of colon cancer. We have found that CFTR and AF-6/afadin are co-localized at the cell-cell contacts and physically interact with each other in colon cancer cell lines. Knockdown of CFTR results in reduced epithelial tightness and enhanced malignancies, with increased degradation and reduced stability of AF-6/afadin protein. The enhanced invasive phenotype of CFTR-knockdown cells can be completely reversed by either AF-6/afadin over-expression or ERK inhibitor, indicating the involvement of AF-6/MAPK pathway. More interestingly, the expression levels of CFTR and AF-6/afadin are significantly downregulated in human colon cancer tissues and lower expression of CFTR and/or AF-6/afadin is correlated with poor prognosis of colon cancer patients. The present study has revealed a previously unrecognized interaction between CFTR and AF-6/afadin that is involved in the pathogenesis of colon cancer and indicated the potential of the two as novel markers of metastasis and prognostic predictors for human colon cancer.
Collapse
|
169
|
Amsellem V, Dryden NH, Martinelli R, Gavins F, Almagro LO, Birdsey GM, Haskard DO, Mason JC, Turowski P, Randi AM. ICAM-2 regulates vascular permeability and N-cadherin localization through ezrin-radixin-moesin (ERM) proteins and Rac-1 signalling. Cell Commun Signal 2014; 12:12. [PMID: 24593809 PMCID: PMC4015342 DOI: 10.1186/1478-811x-12-12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023] Open
Abstract
Background Endothelial junctions control functions such as permeability, angiogenesis and contact inhibition. VE-Cadherin (VECad) is essential for the maintenance of intercellular contacts. In confluent endothelial monolayers, N-Cadherin (NCad) is mostly expressed on the apical and basal membrane, but in the absence of VECad it localizes at junctions. Both cadherins are required for vascular development. The intercellular adhesion molecule (ICAM)-2, also localized at endothelial junctions, is involved in leukocyte recruitment and angiogenesis. Results In human umbilical vein endothelial cells (HUVEC), both VECad and NCad were found at nascent cell contacts of sub-confluent monolayers, but only VECad localized at the mature junctions of confluent monolayers. Inhibition of ICAM-2 expression by siRNA caused the appearance of small gaps at the junctions and a decrease in NCad junctional staining in sub-confluent monolayers. Endothelioma lines derived from WT or ICAM-2-deficient mice (IC2neg) lacked VECad and failed to form junctions, with loss of contact inhibition. Re-expression of full-length ICAM-2 (IC2 FL) in IC2neg cells restored contact inhibition through recruitment of NCad at the junctions. Mutant ICAM-2 lacking the binding site for ERM proteins (IC2 ΔERM) or the cytoplasmic tail (IC2 ΔTAIL) failed to restore junctions. ICAM-2-dependent Rac-1 activation was also decreased in these mutant cell lines. Barrier function, measured in vitro via transendothelial electrical resistance, was decreased in IC2neg cells, both in resting conditions and after thrombin stimulation. This was dependent on ICAM-2 signalling to the small GTPase Rac-1, since transendothelial electrical resistance of IC2neg cells was restored by constitutively active Rac-1. In vivo, thrombin-induced extravasation of FITC-labeled albumin measured by intravital fluorescence microscopy in the mouse cremaster muscle showed that permeability was increased in ICAM-2-deficient mice compared to controls. Conclusions These results indicate that ICAM-2 regulates endothelial barrier function and permeability through a pathway involving N-Cadherin, ERMs and Rac-1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Anna M Randi
- Imperial College for Translational and Experimental Medicine, NHLI Vascular Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12, ONN, UK.
| |
Collapse
|
170
|
Toyoshima D, Mandai K, Maruo T, Supriyanto I, Togashi H, Inoue T, Mori M, Takai Y. Afadin regulates puncta adherentia junction formation and presynaptic differentiation in hippocampal neurons. PLoS One 2014; 9:e89763. [PMID: 24587018 PMCID: PMC3937348 DOI: 10.1371/journal.pone.0089763] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 01/25/2014] [Indexed: 12/31/2022] Open
Abstract
The formation and remodeling of mossy fiber-CA3 pyramidal cell synapses in the stratum lucidum of the hippocampus are implicated in the cellular basis of learning and memory. Afadin and its binding cell adhesion molecules, nectin-1 and nectin-3, together with N-cadherin, are concentrated at puncta adherentia junctions (PAJs) in these synapses. Here, we investigated the roles of afadin in PAJ formation and presynaptic differentiation in mossy fiber-CA3 pyramidal cell synapses. At these synapses in the mice in which the afadin gene was conditionally inactivated before synaptogenesis by using nestin-Cre mice, the immunofluorescence signals for the PAJ components, nectin-1, nectin-3 and N-cadherin, disappeared almost completely, while those for the presynaptic components, VGLUT1 and bassoon, were markedly decreased. In addition, these signals were significantly decreased in cultured afadin-deficient hippocampal neurons. Furthermore, the interevent interval of miniature excitatory postsynaptic currents was prolonged in the cultured afadin-deficient hippocampal neurons compared with control neurons, indicating that presynaptic functions were suppressed or a number of synapse was reduced in the afadin-deficient neurons. Analyses of presynaptic vesicle recycling and paired recordings revealed that the cultured afadin-deficient neurons showed impaired presynaptic functions. These results indicate that afadin regulates both PAJ formation and presynaptic differentiation in most mossy fiber-CA3 pyramidal cell synapses, while in a considerable population of these neurons, afadin regulates only PAJ formation but not presynaptic differentiation.
Collapse
Affiliation(s)
- Daisaku Toyoshima
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
| | - Kenji Mandai
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
- * E-mail: (YT), (KM)
| | - Tomohiko Maruo
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
| | - Irwan Supriyanto
- Faculty of Health Sciences, Kobe University Graduate School of Health Sciences, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
| | - Hideru Togashi
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
| | - Takahito Inoue
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
| | - Masahiro Mori
- Faculty of Health Sciences, Kobe University Graduate School of Health Sciences, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
| | - Yoshimi Takai
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- CREST, Japan Science and Technology Agency, Kobe, Hyogo, Japan
- * E-mail: (YT), (KM)
| |
Collapse
|
171
|
Sufiawati I, Tugizov SM. HIV-associated disruption of tight and adherens junctions of oral epithelial cells facilitates HSV-1 infection and spread. PLoS One 2014; 9:e88803. [PMID: 24586397 PMCID: PMC3931628 DOI: 10.1371/journal.pone.0088803] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 01/15/2014] [Indexed: 11/18/2022] Open
Abstract
Herpes simplex virus (HSV) types 1 and 2 are the most common opportunistic infections in HIV/AIDS. In these immunocompromised individuals, HSV-1 reactivates and replicates in oral epithelium, leading to oral disorders such as ulcers, gingivitis, and necrotic lesions. Although the increased risk of HSV infection may be mediated in part by HIV-induced immune dysfunction, direct or indirect interactions of HIV and HSV at the molecular level may also play a role. In this report we show that prolonged interaction of the HIV proteins tat and gp120 and cell-free HIV virions with polarized oral epithelial cells leads to disruption of tight and adherens junctions of epithelial cells through the mitogen-activated protein kinase signaling pathway. HIV-induced disruption of oral epithelial junctions facilitates HSV-1 paracellular spread between the epithelial cells. Furthermore, HIV-associated disruption of adherens junctions exposes sequestered nectin-1, an adhesion protein and critical receptor for HSV envelope glycoprotein D (gD). Exposure of nectin-1 facilitates binding of HSV-1 gD, which substantially increases HSV-1 infection of epithelial cells with disrupted junctions over that of cells with intact junctions. Exposed nectin-1 from disrupted adherens junctions also increases the cell-to-cell spread of HSV-1 from infected to uninfected oral epithelial cells. Antibodies to nectin-1 and HSV-1 gD substantially reduce HSV-1 infection and cell-to-cell spread, indicating that HIV-promoted HSV infection and spread are mediated by the interaction of HSV gD with HIV-exposed nectin-1. Our data suggest that HIV-associated disruption of oral epithelial junctions may potentiate HSV-1 infection and its paracellular and cell-to-cell spread within the oral mucosal epithelium. This could be one of the possible mechanisms of rapid development of HSV-associated oral lesions in HIV-infected individuals.
Collapse
Affiliation(s)
- Irna Sufiawati
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Sharof M. Tugizov
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
172
|
Elloul S, Kedrin D, Knoblauch NW, Beck AH, Toker A. The adherens junction protein afadin is an AKT substrate that regulates breast cancer cell migration. Mol Cancer Res 2013; 12:464-76. [PMID: 24269953 DOI: 10.1158/1541-7786.mcr-13-0398] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED The PI3K-AKT signaling pathway regulates all phenotypes that contribute to progression of human cancers, including breast cancer. AKT mediates signal relay by phosphorylating numerous substrates, which are causally implicated in biologic responses such as cell growth, survival, metabolic reprogramming, migration, and invasion. Here a new AKT substrate is identified, the adherens junction protein Afadin, which is phosphorylated by AKT at Ser1718. Importantly, under conditions of physiologic IGF-1 signaling and oncogenic PI3K and AKT, Afadin is phosphorylated by all AKT isoforms, and this phosphorylation elicits a relocalization of Afadin from adherens junctions to the nucleus. Also, phosphorylation of Afadin increased breast cancer cell migration that was dependent on Ser1718 phosphorylation. Finally, nuclear localization of Afadin was observed in clinical breast cancer specimens, indicating that regulation of Afadin by the PI3K-AKT pathway has pathophysiologic significance. IMPLICATIONS Phosphorylation of the adhesion protein Afadin by AKT downstream of the PI3K pathway, leads to redistribution of Afadin and controls cancer cell migration.
Collapse
Affiliation(s)
- Sivan Elloul
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115.
| | | | | | | | | |
Collapse
|
173
|
Abstract
Increased endothelial permeability and reduction of alveolar liquid clearance capacity are two leading pathogenic mechanisms of pulmonary edema, which is a major complication of acute lung injury, severe pneumonia, and acute respiratory distress syndrome, the pathologies characterized by unacceptably high rates of morbidity and mortality. Besides the success in protective ventilation strategies, no efficient pharmacological approaches exist to treat this devastating condition. Understanding of fundamental mechanisms involved in regulation of endothelial permeability is essential for development of barrier protective therapeutic strategies. Ongoing studies characterized specific barrier protective mechanisms and identified intracellular targets directly involved in regulation of endothelial permeability. Growing evidence suggests that, although each protective agonist triggers a unique pattern of signaling pathways, selected common mechanisms contributing to endothelial barrier protection may be shared by different barrier protective agents. Therefore, understanding of basic barrier protective mechanisms in pulmonary endothelium is essential for selection of optimal treatment of pulmonary edema of different etiology. This article focuses on mechanisms of lung vascular permeability, reviews major intracellular signaling cascades involved in endothelial monolayer barrier preservation and summarizes a current knowledge regarding recently identified compounds which either reduce pulmonary endothelial barrier disruption and hyperpermeability, or reverse preexisting lung vascular barrier compromise induced by pathologic insults.
Collapse
Affiliation(s)
- Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA.
| | | | | |
Collapse
|
174
|
Komaki R, Togashi H, Takai Y. Regulation of dendritic filopodial interactions by ZO-1 and implications for dendrite morphogenesis. PLoS One 2013; 8:e76201. [PMID: 24098443 PMCID: PMC3788765 DOI: 10.1371/journal.pone.0076201] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 08/21/2013] [Indexed: 12/03/2022] Open
Abstract
Neuronal dendrites dynamically protrude many fine filopodia in the early stages of neuronal development and gradually establish complex structures. The importance of the dendritic filopodia in the formation of axo-dendritic connections is established, but their role in dendrite morphogenesis remains unknown. Using time-lapse imaging of cultured rat hippocampal neurons, we revealed here that many filopodia dynamically protruded from dendrites and transiently interacted with each other to form dendritic filopodia-filopodia contacts in the early stages of neuronal development. The MAGUK family member, Zonula Occludens-1 (ZO-1), which is known to be associated with the nectin and cadherin cell adhesion systems, was concentrated at these dendritic filopodia-filopodia contact sites and also at the tips of free dendritic filopodia. Overexpression of ZO-1 increased the formation of dendritic filopodia and their interactions, and induced abnormal dendrite morphology. Conversely, knockdown of ZO-1 decreased the formation of dendritic filopodia and their interactions, and induced abnormal dendrite morphology which was different from that induced by the overexpression of ZO-1. The components of the nectin and cadherin systems were co-localized with ZO-1 at the dendritic filopodia-filopodia contact sites, but not at the tips of free dendritic filopodia. Overexpression of ZO-1 increased the accumulation of these cell adhesive components at the dendritic filopodia-filopodia contact sites and stabilized their interactions, whereas knockdown of ZO-1 reduced their accumulation at the dendritic filopodia-filopodia contact sites. These results indicate that ZO-1 regulates dendritic filopodial dynamics, which is implicated in dendrite morphogenesis cooperatively with the nectin and cadherin systems in cultured neurons.
Collapse
Affiliation(s)
- Ryouhei Komaki
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideru Togashi
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshimi Takai
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- CREST, Japan Science and Technology Agency, Kobe, Japan
- * E-mail:
| |
Collapse
|
175
|
Ivanov AI, Naydenov NG. Dynamics and regulation of epithelial adherens junctions: recent discoveries and controversies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 303:27-99. [PMID: 23445808 DOI: 10.1016/b978-0-12-407697-6.00002-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Adherens junctions (AJs) are evolutionarily conserved plasma-membrane structures that mediate cell-cell adhesions in multicellular organisms. They are organized by several types of adhesive integral membrane proteins, most notably cadherins and nectins that are clustered and stabilized by a number of cytoplasmic scaffolds. AJs are key regulators of tissue architecture and dynamics via control of cell proliferation, polarity, shape, motility, and survival. They are absolutely critical for normal tissue morphogenesis and their disruption results in pathological abnormalities in different tissues. Although the field of adherens-junction research dramatically progressed in recent years, a number of important questions remain controversial and poorly understood. This review outlines basic principles that regulate organization of AJs in mammalian epithelia and discusses recent advances and standing controversies in the field. A special attention is paid to the regulation of AJs by vesicle trafficking and the intracellular cytoskeleton as well as roles and mechanisms of adherens-junction disruption during tumor progression and tissue inflammation.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA.
| | | |
Collapse
|
176
|
Birukova AA, Tian X, Tian Y, Higginbotham K, Birukov KG. Rap-afadin axis in control of Rho signaling and endothelial barrier recovery. Mol Biol Cell 2013; 24:2678-88. [PMID: 23864716 PMCID: PMC3756920 DOI: 10.1091/mbc.e13-02-0098] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
This study describes the novel role of Rap1 as a molecular switch for down-regulation of the Rho-dependent pathway of agonist-induced endothelial hyperpermeability. The Rho-Rap-Rac autoregulation loop may represent a fundamental mechanism of homeostasis and be critical for reestablishment of cell monolayer integrity in pathological conditions. Activation of the Rho GTPase pathway determines endothelial cell (EC) hyperpermeability after injurious stimuli. To date, feedback mechanisms of Rho down-regulation critical for barrier restoration remain poorly understood. We tested a hypothesis that Rho down-regulation and barrier recovery of agonist-stimulated ECs is mediated by the Ras family GTPase Rap1. Thrombin-induced EC permeability driven by rapid activation of the Rho GTPase pathway was followed by Src kinase–dependent phosphorylation of the Rap1-specific guanine nucleotide exchange factor (GEF) C3G, activation of Rap1, and initiation of EC barrier recovery. Knockdown experiments showed that Rap1 activation was essential for down-regulation of Rho signaling and actin stress fiber dissolution. Rap1 activation also enhanced interaction between adherens junction (AJ) proteins VE-cadherin and p120-catenin and stimulated AJ reannealing mediated by the Rap1 effector afadin. This mechanism also included Rap1-dependent membrane translocation of the Rac1-specific GEF Tiam1 and activation of Rac1-dependent peripheral cytoskeletal dynamics, leading to resealing of intercellular gaps. These data demonstrate that activation of the Rap1-afadin axis is a physiological mechanism driving restoration of barrier integrity in agonist-stimulated EC monolayers via negative-feedback regulation of Rho signaling, stimulation of actin peripheral dynamics, and reestablishment of cell–cell adhesive complexes.
Collapse
Affiliation(s)
- Anna A Birukova
- Department of Medicine, Section of Pulmonary and Critical Medicine, Lung Injury Center, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
177
|
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: 78] [Impact Index Per Article: 6.5] [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
|
178
|
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: 255] [Impact Index Per Article: 21.3] [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
|
179
|
Huveneers S, de Rooij J. Mechanosensitive systems at the cadherin-F-actin interface. J Cell Sci 2013; 126:403-13. [PMID: 23524998 DOI: 10.1242/jcs.109447] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cells integrate biochemical and mechanical information to function within multicellular tissue. Within developing and remodeling tissues, mechanical forces contain instructive information that governs important cellular processes that include stem cell maintenance, differentiation and growth. Although the principles of signal transduction (protein phosphorylation, allosteric regulation of enzymatic activity and binding sites) are the same for biochemical and mechanical-induced signaling, the first step of mechanosensing, in which protein complexes under tension transduce changes in physical force into cellular signaling, is very different, and the molecular mechanisms are only beginning to be elucidated. In this Commentary, we focus on mechanotransduction at cell-cell junctions, aiming to comprehend the molecular mechanisms involved. We describe how different junction structures are associated with the actomyosin cytoskeleton and how this relates to the magnitude and direction of forces at cell-cell junctions. We discuss which cell-cell adhesion receptors have been shown to take part in mechanotransduction. Then we outline the force-induced molecular events that might occur within a key mechanosensitive system at cell-cell junctions; the cadherin-F-actin interface, at which α-catenin and vinculin form a central module. Mechanotransduction at cell-cell junctions emerges as an important signaling mechanism, and we present examples of its potential relevance for tissue development and disease.
Collapse
Affiliation(s)
- Stephan Huveneers
- Sanquin Research and Swammerdam Institute for Life Sciences, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
| | | |
Collapse
|
180
|
Herr D, Fraser HM, Konrad R, Holzheu I, Kreienberg R, Wulff C. Human chorionic gonadotropin controls luteal vascular permeability via vascular endothelial growth factor by down-regulation of a cascade of adhesion proteins. Fertil Steril 2013; 99:1749-58. [PMID: 23465821 DOI: 10.1016/j.fertnstert.2013.01.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 01/07/2013] [Accepted: 01/17/2013] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To study the functional interactions of junctional proteins acting as regulators of vascular permeability in the human corpus luteum. We investigated the role of vascular endothelial (VE)-cadherin, nectin 2, and claudin 5 as controllers of vascular endothelial cell permeability. DESIGN Performing immunohistochemical dual staining, we colocalized the above-mentioned proteins in the human corpus luteum. SETTING Not applicable. PATIENT(S) Not applicable. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) Using a granulosa-endothelial coculture system, we revealed that hCG-treatment down-regulates VE-cadherin, nectin 2, and claudin 5 in endothelial cells via vascular endothelial growth factor (VEGFA). RESULT(S) Furthermore, the interaction of VE-cadherin, nectin 2, and claudin 5 was investigated by silencing these proteins that perform siRNA knockdown. Interestingly, knockdown of VE-cadherin and claudin 5 induced a decrease of the respective other protein. This down-regulation was associated with changed rates of vascular permeability: hCG induced a VEGFA-dependent down-regulation of VE-cadherin, nectin 2, and claudin 5, which increased the endothelial permeability in the coculture system. Furthermore, knockdown of VE-cadherin, nectin-2, and claudin 5 also resulted in a consecutive increase of endothelial permeability for each different protein. CONCLUSION(S) These results demonstrate for the first time that VE-cadherin, nectin 2, and claudin 5 are involved in the regulation of vascular permeability in a mutually interacting manner, which indicates their prominent role for the functionality of the human corpus luteum.
Collapse
Affiliation(s)
- Daniel Herr
- Department of Obstetrics and Gynaecology, Ulm University Medical Centre, Ulm, Germany
| | | | | | | | | | | |
Collapse
|
181
|
Lee KJ, Queenan BN, Rozeboom AM, Bellmore R, Lim ST, Vicini S, Pak DTS. Mossy fiber-CA3 synapses mediate homeostatic plasticity in mature hippocampal neurons. Neuron 2013; 77:99-114. [PMID: 23312519 DOI: 10.1016/j.neuron.2012.10.033] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2012] [Indexed: 11/19/2022]
Abstract
Network activity homeostatically alters synaptic efficacy to constrain neuronal output. However, it is unclear how such compensatory adaptations coexist with synaptic information storage, especially in established networks. Here, we report that in mature hippocampal neurons in vitro, network activity preferentially regulated excitatory synapses within the proximal dendrites of CA3 neurons. These homeostatic synapses exhibited morphological, functional, and molecular signatures of the specialized contacts between mossy fibers of dentate granule cells and thorny excrescences (TEs) of CA3 pyramidal neurons. In vivo TEs were also selectively and bidirectionally altered by chronic activity changes. TE formation required presynaptic synaptoporin and was suppressed by the activity-inducible kinase, Plk2. These results implicate the mossy fiber-TE synapse as an independently tunable gain control locus that permits efficacious homeostatic adjustment of mossy fiber-CA3 synapses, while preserving synaptic weights that may encode information elsewhere within the mature hippocampal circuit.
Collapse
Affiliation(s)
- Kea Joo Lee
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | | | | | | | |
Collapse
|
182
|
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]
|
183
|
Zhang X, Lu G, Qi J, Li Y, He Y, Xu X, Shi J, Zhang CWH, Yan J, Gao GF. Structure of measles virus hemagglutinin bound to its epithelial receptor nectin-4. Nat Struct Mol Biol 2013; 20:67-72. [PMID: 23202587 DOI: 10.1038/nsmb.2432] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 10/01/2012] [Indexed: 12/14/2022]
Abstract
Measles virus is a major public health concern worldwide. Three measles virus cell receptors have been identified so far, and the structures of the first two in complex with measles virus hemagglutinin (MV-H) have been reported. Nectin-4 is the most recently identified receptor in epithelial cells, and its binding mode to MV-H remains elusive. In this study, we solved the structure of the membrane-distal domain of human nectin-4 in complex with MV-H. The structure shows that nectin-4 binds the MV-H β4-β5 groove exclusively via its N-terminal IgV domain; the contact interface is dominated by hydrophobic interactions. The binding site in MV-H for nectin-4 also overlaps extensively with those of the other two receptors. Finally, a hydrophobic pocket centered in the β4-β5 groove is involved in binding to all three identified measles virus receptors, representing a potential target for antiviral drugs.
Collapse
Affiliation(s)
- Xiaoai Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Rehm K, Panzer L, van Vliet V, Genot E, Linder S. Drebrin preserves endothelial integrity by stabilizing nectin at adherens junctions. J Cell Sci 2013; 126:3756-69. [DOI: 10.1242/jcs.129437] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Regulation of cell-cell contacts is essential for integrity of the vascular endothelium. Here, a critical role of the F-actin binding protein drebrin in maintaining endothelial integrity is revealed under conditions mimicking vascular flow. Drebrin knockdown leads to weakening of cell-cell contacts, characterized by loss of nectin from adherens junctions and its subsequent lysosomal degradation. Immunoprecipitation, FRAP and mitochondrial retargeting experiments show that nectin stabilization occurs through a chain of interactions: drebrin binding to F-actin, interaction of drebrin and afadin through their polyproline and PR1-2 regions, and recruitment of nectin through afadin's PDZ region. Key elements are drebrin's modules that confer binding to afadin and F-actin. Evidence is provided by constructs containing afadin's PDZ region coupled to drebrin's F-actin binding region or to lifeact, which restore junctional nectin under knockdown of drebrin or of both drebrin and afadin. Drebrin, containing binding sites for both afadin and F-actin, is thus uniquely equipped to stabilize nectin at endothelial junctions and to preserve endothelial integrity under vascular flow.
Collapse
|
185
|
Insights into the role of cell-cell junctions in physiology and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 306:187-221. [PMID: 24016526 DOI: 10.1016/b978-0-12-407694-5.00005-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Contacting cells establish different classes of intricate structures at the cell-cell junctions. These structures are of increasing research interest as they regulate a broad variety of processes in development and disease. Further, in vitro studies are revealing that various cell-cell interaction proteins are involved not only in cell-cell processes but also in many additional aspects of physiology, such as migration and apoptosis. This chapter reviews the basic classification of cell-cell junctional structures and some of their representative proteins. Their roles in development and disease are briefly outlined, followed by a section on contemporary methods for probing cell-cell interactions and some recent developments. This chapter concludes with a few suggestions for potential research directions to further develop this promising area of study.
Collapse
|
186
|
Xiao X, Cheng CY, Mruk DD. Intercellular adhesion molecule-2 is involved in apical ectoplasmic specialization dynamics during spermatogenesis in the rat. J Endocrinol 2013; 216:73-86. [PMID: 23097088 PMCID: PMC3535886 DOI: 10.1530/joe-12-0434] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this study, we investigated the role of intercellular adhesion molecule-2 (ICAM2) in the testis. ICAM2 is a cell adhesion protein having important roles in cell migration, especially during inflammation when leukocytes cross the endothelium. Herein, we showed ICAM2 to be expressed by germ and Sertoli cells in the rat testis. When a monospecific antibody was used for immunolocalization experiments, ICAM2 was found to surround the heads of elongating/elongated spermatids in all stages of the seminiferous epithelial cycle. To determine whether ICAM2 is a constituent of apical ectoplasmic specialization (ES), co-immunoprecipitation and dual immunofluorescence staining were performed. Interestingly, ICAM2 was found to associate with β1-integrin, nectin-3, afadin, Src, proline-rich tyrosine kinase 2, annexin II, and actin. Following CdCl₂ treatment, ICAM2 was found to be upregulated during restructuring of the seminiferous epithelium, with round spermatids becoming increasingly immunoreactive for ICAM2 by 6-16 h. Interestingly, there was a loss in the binding of ICAM2 to actin during CdCl₂-induced germ cell loss, suggesting that a loss of ICAM2-actin interactions might have facilitated junction restructuring. Taken collectively, these results illustrate that ICAM2 plays an important role in apical ES dynamics during spermatogenesis.
Collapse
|
187
|
Monteiro AC, Parkos CA. Intracellular mediators of JAM-A-dependent epithelial barrier function. Ann N Y Acad Sci 2012; 1257:115-24. [PMID: 22671597 DOI: 10.1111/j.1749-6632.2012.06521.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Junctional adhesion molecule-A (JAM-A) is a critical signaling component of the apical junctional complex, a structure composed of several transmembrane and scaffold molecules that controls the passage of nutrients and solutes across epithelial surfaces. Observations from JAM-A-deficient epithelial cells and JAM-A knockout animals indicate that JAM-A is an important regulator of epithelial paracellular permeability; however, the mechanism(s) linking JAM-A to barrier function are not understood. This review highlights recent findings relevant to JAM-A-mediated regulation of epithelial permeability, focusing on the role of upstream and downstream signaling candidates. We draw on what is known about proteins reported to associate with JAM-A in other pathways and on known modulators of barrier function to propose candidate effectors that may mediate JAM-A regulation of epithelial paracellular permeability. Further investigation of pathways highlighted in this review may provide ideas for novel therapeutics that target debilitating conditions associated with barrier dysfunction, such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Ana C Monteiro
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology Research Unit, Emory University, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
188
|
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
|
189
|
Schust DJ, Ibana JA, Buckner LR, Ficarra M, Sugimoto J, Amedee AM, Quayle AJ. Potential mechanisms for increased HIV-1 transmission across the endocervical epithelium during C. trachomatis infection. Curr HIV Res 2012; 10:218-27. [PMID: 22384841 DOI: 10.2174/157016212800618093] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/18/2011] [Accepted: 12/28/2011] [Indexed: 11/22/2022]
Abstract
Among the now pandemic sexually transmitted infections (STIs), Chlamydia trachomatis (C. trachomatis) is the predominant bacterial pathogen and human immunodeficiency virus type 1 (HIV-1) is the most lethal of the viral pathogens. The female genital tract is the primary site for heterosexual transmission of both C. trachomatis and HIV-1. Infection with C. trachomatis, and with a variety of other STIs, increases the risk for transmission of HIV-1, although the mechanisms for this finding remain unclear. We have used in vitro modeling to assess the mechanisms by which infection with genital C. trachomatis serovars might increase the transmission of HIV-1 across the female genital tract. C. trachomatis infection of an immortalized endocervical epithelial cell line (A2EN) increases the cell surface expression of the HIV-1 alternative primary receptor, galactosyl ceramide (GalCer), and of the HIV-1 co-receptors, CXCR4 and CCR5. C. trachomatis infection also increases the binding of HIV-1 to A2EN cells, and, subsequently, increases levels of virus in co-cultures of HIV-exposed A2EN and susceptible MT4-R5 T cells. Finally, in vivo endocervical cell sampling reveals a dramatic increase in the number of CD4+, CXCR4 and/or CCR5 positive T cell targets in the endocervix of C. trachomatis positive women when compared to those who are C. trachomatis negative. This combination of in vitro and in vivo results suggests several mechanisms for increased transmission of HIV-1 across the endocervices of C. trachomatis-infected women.
Collapse
Affiliation(s)
- Danny J Schust
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA.
| | | | | | | | | | | | | |
Collapse
|
190
|
Bojesen KB, Clausen O, Rohde K, Christensen C, Zhang L, Li S, Køhler L, Nielbo S, Nielsen J, Gjørlund MD, Poulsen FM, Bock E, Berezin V. Nectin-1 binds and signals through the fibroblast growth factor receptor. J Biol Chem 2012; 287:37420-33. [PMID: 22955284 DOI: 10.1074/jbc.m112.345215] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Nectins belong to a family of immunoglobulin (Ig)-like cell-adhesion molecules comprising four members, nectin-1 through nectin-4. Nectins are involved in formation of the mechanical adhesive puncta adherentia junctions of synapses. Nectins share the same overall structural topology with an extracellular region containing three Ig modules, a transmembrane region, and a cytoplasmic region. In nectin-1, the first and second Ig module in the extracellular region are necessary for the trans-interaction with nectin-3 and formation of cis-dimers, respectively. The function of the third Ig module of nectin-1 remains unknown. We here report the structure in solution of the third, membrane-proximal Ig module of mouse nectin-1 (nectin-1 Ig3) solved by means of nuclear magnetic resonance (NMR) spectroscopy. It belongs to the C1 set of the Ig superfamily. Nectin-1 Ig3 was produced as a recombinant protein and induced neurite outgrowth in primary cultures of hippocampal and cerebellar granule neurons, an effect abolished by treatment with the fibroblast growth factor receptor (FGFR) inhibitor SU5402, or by transfection with a dominant-negative FGFR1 construct. We showed by surface plasmon resonance (SPR) analysis that nectin-1 Ig3 directly interacted with various isoforms of FGFR. Nectin-1 Ig3 induced phosphorylation of FGFR1c in the same manner as the whole nectin-1 ectodomain, and promoted survival of cerebellar granule neurons induced to undergo apoptosis. Finally, we constructed a peptide, nectide, by employing in silico modeling of various FGFR ligand-binding sites. Nectide mimicked all the effects of nectin-1 Ig3. We suggest that FGFR is a downstream signaling partner of nectin-1.
Collapse
Affiliation(s)
- Kirsten B Bojesen
- Protein Laboratory, Department of Neuroscience and Pharmacology, Panum Institute, Blegdamsvej 3C, DK-2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Ballester M, Gonin J, Rodenas A, Bernaudin JF, Rouzier R, Coutant C, Daraï E. Eutopic endometrium and peritoneal, ovarian and colorectal endometriotic tissues express a different profile of Nectin-1, -3, -4 and nectin-like molecule 2. Hum Reprod 2012; 27:3179-86. [DOI: 10.1093/humrep/des304] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
192
|
Harrison OJ, Vendome J, Brasch J, Jin X, Hong S, Katsamba PS, Ahlsen G, Troyanovsky RB, Troyanovsky SM, Honig B, Shapiro L. Nectin ectodomain structures reveal a canonical adhesive interface. Nat Struct Mol Biol 2012; 19:906-15. [PMID: 22902367 PMCID: PMC3443293 DOI: 10.1038/nsmb.2366] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/19/2012] [Indexed: 11/21/2022]
Abstract
Nectins are immunoglobulin superfamily glycoproteins that mediate intercellular adhesion in many vertebrate tissues. Homophilic and heterophilic interactions between nectin family members help to mediate tissue patterning. We determined homophilic binding affinities and heterophilic specificities of all four nectins and the related protein nectin-like 5 from human and mouse, revealing a range of homophilic strengths and a defined heterophilic specificity pattern. To understand the molecular basis of adhesion and specificity, we determined crystal structures of natively glycosylated full ectodomains or adhesive fragments of nectins 1–4 and nectin-like 5. All crystal structures reveal dimeric nectins bound through a stereotyped interface previously proposed to represent a cis dimer. However, conservation of this interface and results of targeted cross-linking experiments show that this dimer likely represents the adhesive trans interaction. Its structure provides a simple molecular explanation for the adhesive binding specificity of nectins.
Collapse
Affiliation(s)
- Oliver J Harrison
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Xu X, Zhang X, Lu G, Cai Y. Purification, crystallization and preliminary X-ray analysis of the IgV domain of human nectin-4. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:942-5. [PMID: 22869128 PMCID: PMC3412779 DOI: 10.1107/s1744309112027236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/15/2012] [Indexed: 12/18/2022]
Abstract
Nectin-4 belongs to a family of immunoglobulin-like cell adhesion molecules and is highly expressed in cancer cells. Recently, nectin-4 was found to be a receptor of measles virus and the IgV domain sustains strong binding to measles virus H protein. In this study, the successful expression and purification of human nectin-4 V domain (nectin-4v) is reported. The purified protein was crystallized using the sitting-drop vapour-diffusion method. The crystals diffracted to 1.8 Å resolution and belonged to space group P2(1), with unit-cell parameters a = 33.1, b = 51.7, c = 56.9 Å, β = 94.7°. Preliminary analysis of the diffraction data was also performed.
Collapse
Affiliation(s)
- Xiang Xu
- College of Life Science, Anhui Agricultural University, Anhui 230036, People’s Republic of China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People’s Republic of China
| | - Xiaoai Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People’s Republic of China
| | - Guangwen Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People’s Republic of China
| | - Yongping Cai
- College of Life Science, Anhui Agricultural University, Anhui 230036, People’s Republic of China
| |
Collapse
|
194
|
Sadayappan S, de Tombe PP. Cardiac myosin binding protein-C: redefining its structure and function. Biophys Rev 2012; 4:93-106. [PMID: 22707987 PMCID: PMC3374655 DOI: 10.1007/s12551-012-0067-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/13/2012] [Indexed: 01/10/2023] Open
Abstract
Mutations of cardiac myosin binding protein-C (cMyBP-C) are inherited by an estimated 60 million people worldwide, and the protein is the target of several kinases. Recent evidence further suggests that cMyBP-C mutations alter Ca(2+) transients, leading to electrophysiological dysfunction. Thus, while the importance of studying this cardiac sarcomere protein is clear, preliminary data in the literature have raised many questions. Therefore, in this article, we propose to review the structure and function of cMyBP-C with particular respect to the role(s) in cardiac contractility and whether its release into the circulatory system is a potential biomarker of myocardial infarction. We also discuss future directions and experimental designs that may lead to expanding the role(s) of cMyBP-C in the heart. In conclusion, we suggest that cMyBP-C is a regulatory protein that could offer a broad clinical utility in maintaining normal cardiac function.
Collapse
Affiliation(s)
- Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave., Maywood, IL 60153 USA
| | - Pieter P. de Tombe
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, 2160 South First Ave., Maywood, IL 60153 USA
| |
Collapse
|
195
|
Maniwa Y, Nishio W, Okita Y, Yoshimura M. Expression of nectin 3: Novel prognostic marker of lung adenocarcinoma. Thorac Cancer 2012; 3:175-181. [DOI: 10.1111/j.1759-7714.2011.00104.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
196
|
Birukova AA, Fu P, Wu T, Dubrovskyi O, Sarich N, Poroyko V, Birukov KG. Afadin controls p120-catenin-ZO-1 interactions leading to endothelial barrier enhancement by oxidized phospholipids. J Cell Physiol 2012; 227:1883-90. [PMID: 21732359 DOI: 10.1002/jcp.22916] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Afadin is a novel regulator of epithelial cell junctions assembly. However, its role in the formation of endothelial cell junctions and the regulation of vascular permeability remains obscure. We previously described protective effects of oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) in the in vitro and in vivo models of lung endothelial barrier dysfunction and acute lung injury, which were mediated by Rac GTPase. This study examined a role of afadin in the OxPAPC-induced enhancement of interactions between adherens junctions and tight junctions as a novel mechanism of endothelial cell (EC) barrier preservation. OxPAPC induced Rap1-dependent afadin accumulation at the cell periphery and Rap1-dependent afadin interaction with adherens junction and tight junction proteins p120-catenin and ZO-1, respectively. Afadin knockdown using siRNA or ectopic expression of afadin mutant lacking Rap1 GTPase binding domain suppressed OxPAPC-induced EC barrier enhancement and abolished barrier protective effects of OxPAPC against thrombin-induced EC permeability. Afadin knockdown also abolished protective effects of OxPAPC against ventilator-induced lung injury in vivo. These results demonstrate for the first time a critical role of afadin in the regulation of vascular barrier function in vitro and in vivo via coordination of adherens junction-tight junction interactions.
Collapse
Affiliation(s)
- Anna A Birukova
- Lung Injury Center, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
197
|
Castellanos KJ, Gagyi E, Kormos B, Valyi-Nagy K, Voros A, Shukla D, Horvath S, Slavin KV, Valyi-Nagy T. Increased axonal expression of nectin-1 in multiple sclerosis plaques. Neurol Sci 2012; 34:465-9. [PMID: 22460696 DOI: 10.1007/s10072-012-1026-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 03/18/2012] [Indexed: 12/26/2022]
Abstract
Nectin-1 is a cell adhesion molecule that plays a role in interneuronal synapse formation, in axonal guidance during development and possibly in neuron-glia interactions. To better understand axonal changes in MS, nectin-1 expression was determined by immunohistochemistry in normal adult human cerebral white matter (n = 4) and in six MS plaques (three active and three inactive). The intensity of axonal nectin-1 expression was scored on a scale of 0 to 4+. In normal adult cerebral white matter, axons showed weak nectin-1 expression with a score of 1.25 ± 0.50. Axonal nectin-1 expression was significantly stronger within both active (score = 3.33 ± 0.289, p = 0.001) and inactive (score = 2.16 ± 0.29, p = 0.038) MS plaques than in normal white matter. Axons in white matter adjacent to MS plaques showed nectin-1 expression (score = 1.5 ± 0.50) that was not statistically different from normal controls (p = 0.542). These findings raise the possibility that increased expression of nectin-1 in MS lesions plays a role in the pathogenesis of MS through participation in axonal responses to injury and mediation of altered neuron-glia interactions relevant to myelination.
Collapse
Affiliation(s)
- Karla J Castellanos
- Department of Pathology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Baek H, Kim JH, Noh YT, Kwon H. The soluble amino-terminal region of HVEM mediates efficient herpes simplex virus type 1 infection of gD receptor-negative cells. Virol J 2012; 9:15. [PMID: 22239829 DOI: 10.1186/1743-422x-9-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 01/13/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Previous studies from our own and other labs reported the surprising finding that the soluble V domain of the herpes simplex virus type 1 (HSV-1) entry receptor nectin-1 can both block HSV infection of receptor-bearing cells and mediate infection of receptor-deficient cells. Here we show that this property is not unique to nectin-1. We generated a pair of truncated, soluble forms of the other major HSV-1 entry receptor, herpes virus entry mediator (HVEM or HveA), and examined its effects on HSV-1 infection of receptor-deficient cells. RESULTS In cultures of CHO-K1 cells, sHveA102 comprising the two amino-terminal cysteine-rich pseudorepeats (CRPs) of HVEM enabled infection of greater than 80% of the cells at an MOI of 3, while sHveA162 comprising the complete ectodomain failed to mediate infection. Both sHveA102 and sHveA162 blocked infection of CHO-K1 cells stably expressing HVEM in a dose-dependent manner, indicating that both were capable of binding to viral gD. We found that sHveA102-mediated infection involves pH-independent endocytosis whereas HSV infection of HVEM-expressing CHO-K1 cells is known to be pH-dependent. CONCLUSIONS Our results suggest that the C-terminal portion of the soluble HVEM ectodomain inhibits gD activation and that this effect is neutralized in the full-length form of HVEM in normal infection.
Collapse
Affiliation(s)
- Hyunjung Baek
- Division of Radiation Oncology, Korea Institute of Radiological and Medical Sciences, 215-4, Gongneung-Dong, Nowon-Ku, Seoul 139-706, South Korea
| | | | | | | |
Collapse
|
199
|
Bonazzi M, Cossart P. Impenetrable barriers or entry portals? The role of cell-cell adhesion during infection. ACTA ACUST UNITED AC 2012; 195:349-58. [PMID: 22042617 PMCID: PMC3206337 DOI: 10.1083/jcb.201106011] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell-cell adhesion plays a fundamental role in cell polarity and organogenesis. It also contributes to the formation and establishment of physical barriers against microbial infections. However, a large number of pathogens, from viruses to bacteria and parasites, have developed countless strategies to specifically target cell adhesion molecules in order to adhere to and invade epithelial cells, disrupt epithelial integrity, and access deeper tissues for dissemination. The study of all these processes has contributed to the characterization of molecular machineries at the junctions of eukaryotic cells that have been better understood by using pathogens as probes.
Collapse
Affiliation(s)
- Matteo Bonazzi
- Centre Nationale de la Recherche Scientifique, UMR 5236, CPBS, CNRS, 34293 Montpellier, France.
| | | |
Collapse
|
200
|
Abstract
Epithelia are highly organised structures protecting underlying tissues against microbial pathogens. Epithelial morphogenesis and maintenance is mediated by cell-cell adhesion molecules organised in junctional complexes, such as the adherens junctions. The tight organisation of these complexes and their interactions with cellular factors render the epithelia impermeable to potential invaders. Nevertheless, pathogens have developed strategies to target, interact and manipulate junctional complexes, in order to disrupt or cross the epithelial barriers and cause infection. Bacteria, viruses and parasites access the junctional molecular components either directly, often taking advantage of physiological alterations in epithelial polarity, or indirectly, by delivering into cells molecular factors that destabilise junctional integrity. Importantly, microbial interactions with junctional components are instrumental not only to elucidate mechanisms of invasion, but also to unravel fundamental physiological properties of the epithelial barriers, at the cellular and tissular level.
Collapse
Affiliation(s)
- Georgios Nikitas
- Biomedical Research Foundation Academy of Athens, 11527, Athens, Greece
| | | |
Collapse
|