1
|
Pelz L, Dossou L, Kompier N, Jüttner R, Siemonsmeier G, Meyer N, Lowenstein ED, Lahmann I, Kettenmann H, Birchmeier C, Rathjen FG. The IgCAM BT-IgSF (IgSF11) is essential for connexin43-mediated astrocyte-astrocyte coupling in mice. eNeuro 2024; 11:ENEURO.0283-23.2024. [PMID: 38388443 PMCID: PMC10957231 DOI: 10.1523/eneuro.0283-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
The type I transmembrane protein BT-IgSF is predominantly localized in the brain and testes. It belongs to the CAR subgroup of Ig cell adhesion proteins, that are hypothesized to regulate connexin expression or localization. Here, we studied the putative link between BT-IgSF and connexins in astrocytes, ependymal cells and neurons of the mouse. Global knockout of BT-IgSF caused an increase in the clustering of connexin43 (Gja1), but not of connexin30 (Gjb6), on astrocytes and ependymal cells. Additionally, knockout animals displayed reduced expression levels of connexin43 protein in the cortex and hippocampus. Importantly, analysis of biocytin spread in hippocampal or cortical slices from mature mice of either sex revealed a decrease in astrocytic cell-cell coupling in the absence of BT-IgSF. Blocking either protein biosynthesis or proteolysis showed that the lysosomal pathway increased connexin43 degradation in astrocytes. Localization of connexin43 in subcellular compartments was not impaired in astrocytes of BT-IgSF mutants. In contrast to connexin43 the localization and expression of connexin36 (Gjd2) on neurons was not affected by the absence of BT-IgSF. Overall, our data indicate that the IgCAM BT-IgSF is essential for correct gap junction-mediated astrocyte-to-astrocyte cell communication.Significance Statement Astrocytes regulate a variety of physiological processes in the developing and adult brain that are essential for proper brain function. Astrocytes form extensive networks in the brain and communicate via gap junctions. Disruptions of gap junction coupling are found in several diseases such as neurodegeneration or epilepsy. Here, we demonstrate that the cell adhesion protein BT-IgSF is essential for gap junction mediated coupling between astrocytes in the cortex and hippocampus.
Collapse
Affiliation(s)
- Laura Pelz
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - Laura Dossou
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - Nine Kompier
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - René Jüttner
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | | | - Niklas Meyer
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | | | - Ines Lahmann
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| | - Helmut Kettenmann
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Carmen Birchmeier
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Fritz G. Rathjen
- Max-Delbrück-Center for Molecular Medicine, Berlin DE-13092, Germany
| |
Collapse
|
2
|
Freiberg F, Thakkar M, Hamann W, Lopez Carballo J, Jüttner R, Voss FK, Becher PM, Westermann D, Tschöpe C, Heuser A, Rocks O, Fischer R, Gotthardt M. CAR links hypoxia signaling to improved survival after myocardial infarction. Exp Mol Med 2023; 55:643-652. [PMID: 36941462 PMCID: PMC10073142 DOI: 10.1038/s12276-023-00963-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/08/2022] [Accepted: 12/25/2022] [Indexed: 03/23/2023] Open
Abstract
The coxsackievirus and adenovirus receptor (CAR) mediates homo- and heterotopic interactions between neighboring cardiomyocytes at the intercalated disc. CAR is upregulated in the hypoxic areas surrounding myocardial infarction (MI). To elucidate whether CAR contributes to hypoxia signaling and MI pathology, we used a gain- and loss-of-function approach in transfected HEK293 cells, H9c2 cardiomyocytes and CAR knockout mice. CAR overexpression increased RhoA activity, HIF-1α expression and cell death in response to chemical and physical hypoxia. In vivo, we subjected cardiomyocyte-specific CAR knockout (KO) and wild-type mice (WT) to coronary artery ligation. Survival was drastically improved in KO mice with largely preserved cardiac function as determined by echocardiography. Histological analysis revealed a less fibrotic, more compact lesion. Thirty days after MI, there was no compensatory hypertrophy or reduced cardiac output in hearts from CAR KO mice, in contrast to control mice with increased heart weight and reduced ejection fraction as signs of the underlying pathology. Based on these findings, we suggest CAR as a therapeutic target for the improved future treatment or prevention of myocardial infarction.
Collapse
Affiliation(s)
- Fabian Freiberg
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Meghna Thakkar
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Wiebke Hamann
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jacobo Lopez Carballo
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Rene Jüttner
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Felizia K Voss
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Peter M Becher
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Hamburg, Germany
- DZHK Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Dirk Westermann
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Hamburg, Germany
- DZHK Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Carsten Tschöpe
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- BCRT (Berlin-Brandenburg Center for Regenerative Therapies), Berlin, Germany
| | - Arnd Heuser
- Animal Phenotyping, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Rocks
- Spatiotemporal Control of Rho GTPase Signaling, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Robert Fischer
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Gotthardt
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Rathjen FG, Jüttner R. The IgSF Cell Adhesion Protein CLMP and Congenital Short Bowel Syndrome (CSBS). Int J Mol Sci 2023; 24:5719. [PMID: 36982793 PMCID: PMC10056934 DOI: 10.3390/ijms24065719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
The immunoglobulin-like cell adhesion molecule CLMP is a member of the CAR family of cell adhesion proteins and is implicated in human congenital short-bowel syndrome (CSBS). CSBS is a rare but very severe disease for which no cure is currently available. In this review, we compare data from human CSBS patients and a mouse knockout model. These data indicate that CSBS is characterized by a defect in intestinal elongation during embryonic development and impaired peristalsis. The latter is driven by uncoordinated calcium signaling via gap junctions, which is linked to a reduction in connexin43 and 45 levels in the circumferential smooth muscle layer of the intestine. Furthermore, we discuss how mutations in the CLMP gene affect other organs and tissues, including the ureter. Here, the absence of CLMP produces a severe bilateral hydronephrosis-also caused by a reduced level of connexin43 and associated uncoordinated calcium signaling via gap junctions.
Collapse
Affiliation(s)
- Fritz G. Rathjen
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany;
| | | |
Collapse
|
4
|
CXADR: From an Essential Structural Component to a Vital Signaling Mediator in Spermatogenesis. Int J Mol Sci 2023; 24:ijms24021288. [PMID: 36674801 PMCID: PMC9865082 DOI: 10.3390/ijms24021288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Canonical coxsackievirus and adenovirus receptor (CXADR) is a transmembrane component of cell junctions that is crucial for cardiac and testicular functions via its homophilic and heterophilic interaction. CXADR is expressed in both Sertoli cells and germ cells and is localized mainly at the interface between Sertoli-Sertoli cells and Sertoli-germ cells. Knockout of CXADR in mouse Sertoli cells specifically impairs male reproductive functions, including a compromised blood-testis barrier, apoptosis of germ cells, and premature loss of spermatids. Apart from serving as an important component for cell junctions, recent progress has showed the potential roles of CXADR as a signaling mediator in spermatogenesis. This review summarizes current research progress related to the regulation and role of CXADR in spermatogenesis as well as in pathological conditions. We hope this review provides some future directions and a blueprint to promote the further study on the roles of CXADR.
Collapse
|
5
|
Klann PJ, Wang X, Elfert A, Zhang W, Köhler C, Güttsches AK, Jacobsen F, Weyen U, Roos A, Ehrke-Schulz E, Ehrhardt A, Vorgerd M, Bayer W. Seroprevalence of Binding and Neutralizing Antibodies against 39 Human Adenovirus Types in Patients with Neuromuscular Disorders. Viruses 2022; 15:79. [PMID: 36680119 PMCID: PMC9866721 DOI: 10.3390/v15010079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
High pre-existing antibodies against viral vectors reduce their functionality and may lead to adverse complications. To circumvent this problem in future gene therapy approaches, we tested the seroprevalence of a large range of human adenovirus types in patients with neuromuscular disorders (NMDs) to find appropriate viral vector candidates for gene replacement therapy for NMDs. Binding and neutralizing antibodies against 39 human adenovirus types were tested in the sera of 133 patients with NMDs and 76 healthy controls aged 17-92 years. The influence of age, sex, and NMDs on antibody levels was analyzed. The seroprevalence of different adenoviruses in the cohort varied widely. The highest levels of binding antibodies were detected against HAdV-D27, -C1, -D24, -D70, -B14, -C6, -D13, -B34, and -E4, whereas the lowest reactivity was detected against HAdV-F41, -A31, -B11, -D75, -D8, -D65, -D26, -D80, and -D17. The highest neutralizing reactivity was observed against HAdV-B3, -C2, -E4, -C1, -G52, -C5, and -F41, whereas the lowest neutralizing reactivity was observed against HAdV-D74, -B34, -D73, -B37, -D48, -D13, -D75, -D8, -B35, and -B16. We detected no influence of sex and only minor differences between different age groups. Importantly, there were no significant differences between healthy controls and patients with NMDs. Our data show that patients with NMDs have very similar levels of binding and neutralizing antibodies against HAdV compared to healthy individuals, and we identified HAdV-A31, -B16, -B34, -B35, -D8, -D37, -D48, -D73, -D74, -D75, and -D80 as promising candidates for future vector development due to their low binding and neutralizing antibody prevalence.
Collapse
Affiliation(s)
- Patrick Julian Klann
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Xiaoyan Wang
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Anna Elfert
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Medical Education and Research, Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Cornelia Köhler
- Clinics for Pediatrics and Adolescent Medicine, University Hospital Sankt Josef, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Anne-Katrin Güttsches
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Frank Jacobsen
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Ute Weyen
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Andreas Roos
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Eric Ehrke-Schulz
- Virology and Microbiology, Center for Medical Education and Research, Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Medical Education and Research, Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Matthias Vorgerd
- Heimer Institute for Muscle Research, Department of Neurology, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
6
|
Matthaeus C, Jüttner R, Gotthardt M, Rathjen FG. The IgCAM CAR Regulates Gap Junction-Mediated Coupling on Embryonic Cardiomyocytes and Affects Their Beating Frequency. Life (Basel) 2022; 13:14. [PMID: 36675963 PMCID: PMC9866089 DOI: 10.3390/life13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The IgCAM coxsackie-adenovirus receptor (CAR) is essential for embryonic heart development and electrical conduction in the mature heart. However, it is not well-understood how CAR exerts these effects at the cellular level. To address this question, we analyzed the spontaneous beating of cultured embryonic hearts and cardiomyocytes from wild type and CAR knockout (KO) embryos. Surprisingly, in the absence of the CAR, cultured cardiomyocytes showed increased frequencies of beating and calcium cycling. Increased beatings of heart organ cultures were also induced by the application of reagents that bind to the extracellular region of the CAR, such as the adenovirus fiber knob. However, the calcium cycling machinery, including calcium extrusion via SERCA2 and NCX, was not disrupted in CAR KO cells. In contrast, CAR KO cardiomyocytes displayed size increases but decreased in the total numbers of membrane-localized Cx43 clusters. This was accompanied by improved cell-cell coupling between CAR KO cells, as demonstrated by increased intercellular dye diffusion. Our data indicate that the CAR may modulate the localization and oligomerization of Cx43 at the plasma membrane, which could in turn influence electrical propagation between cardiomyocytes via gap junctions.
Collapse
Affiliation(s)
- Claudia Matthaeus
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
- Laboratory of Cellular Biophysics, NHLBI, NIH, 50 South Drive, Building 50 RM 3312, Bethesda, MD 20892, USA
| | - René Jüttner
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
| | - Michael Gotthardt
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
| | - Fritz G. Rathjen
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, DE-13092 Berlin, Germany
| |
Collapse
|
7
|
Human Coxsackie- and adenovirus receptor is a putative target of neutrophil elastase-mediated shedding. Mol Biol Rep 2022; 49:3213-3223. [PMID: 35122600 PMCID: PMC8924087 DOI: 10.1007/s11033-022-07153-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 01/17/2022] [Indexed: 12/04/2022]
Abstract
Background During viral-induced myocarditis, immune cells migrate towards the site of infection and secrete proteases, which in turn can act as sheddases by cleaving extracellular domains of transmembrane proteins. We were interested in the shedding of the Coxsackie- and adenovirus receptor (CAR) that acts as an entry receptor for both eponymous viruses, which cause myocarditis. CAR shedding by secreted immune proteases could result in a favourable outcome of myocarditis as CAR’s extracellular domain would be removed from the cardiomyocytes’ surface leading to decreased susceptibility to ongoing viral infections. Methods and results In this work, matrix metalloproteinases and serine proteinases were screened for their proteolytic activity towards human CAR. Whereas matrix metalloproteinases, proteinase 3, and cathepsin G did not cleave human recombinant CAR or only within long incubation times, neutrophil elastase showed a distinct cleavage pattern of CAR’s extracellular domain that was time- and dose-dependent. Neutrophil elastase cleaves CAR at its membrane-proximal immunoglobulin domain as we determined by nanoLC-MS/MS. Furthermore, neutrophil elastase treatment of cells reduced CAR surface levels as seen by flow cytometry and immunofluorescence microscopy. Conclusions With this study, we show that CAR might be a target for shedding by neutrophil elastase. Supplementary Information The online version contains supplementary material available at 10.1007/s11033-022-07153-2.
Collapse
|
8
|
Gresele P, Momi S, Marcucci R, Ramundo F, De Stefano V, Tripodi A. Interactions of adenoviruses with platelets and coagulation and the vaccine-induced immune thrombotic thrombocytopenia syndrome. Haematologica 2021; 106:3034-3045. [PMID: 34407607 PMCID: PMC8634187 DOI: 10.3324/haematol.2021.279289] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
The COVID-19 pandemic has had a heavy impact on global health and economy and vaccination remains the primary way of controlling the infection. During the ongoing vaccination campaign some unexpected thrombotic events have emerged in subjects who had recently received the AstraZeneca (Vaxzevria) vaccine or the Johnson and Johnson (Janssen) vaccine, two adenovirus vector-based vaccines. Epidemiological studies confirm that the observed/expected ratio of these unusual thromboses is abnormally increased, especially in women in fertile age. The characteristics of this complication, with venous thromboses at unusual sites, most frequently in the cerebral vein sinuses but also in splanchnic vessels, often with multiple associated thromboses, thrombocytopenia, and sometimes disseminated intravascular coagulation, are unique and the time course and tumultuous evolution are suggestive of an acute immunological reaction. Indeed, plateletactivating anti-PF4 antibodies have been detected in a large proportion of the affected patients. Several data suggest that adenoviruses may interact with platelets, the endothelium and the blood coagulation system. Here we review interactions between adenoviral vectors and the hemostatic system that are of possible relevance in vaccine-associated thrombotic thrombocytopenia syndrome. We systematically analyze the clinical data on the reported thrombotic complications of adenovirus-based therapeutics and discuss all the current hypotheses on the mechanisms triggering this novel syndrome. Although, considering current evidence, the benefit of vaccination clearly outweighs the potential risks, it is of paramount importance to fully unravel the mechanisms leading to vaccineassociated thrombotic thrombocytopenia syndrome and to identify prognostic factors through further research.
Collapse
Affiliation(s)
- Paolo Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia.
| | - Stefania Momi
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence; Atherothrombotic Center, AOU Careggi, Florence
| | - Francesco Ramundo
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli - IRCCS - Rome
| | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli - IRCCS - Rome
| | - Armando Tripodi
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thromboses Center, Milan
| |
Collapse
|
9
|
Altered cell and RNA isoform diversity in aging Down syndrome brains. Proc Natl Acad Sci U S A 2021; 118:2114326118. [PMID: 34795060 PMCID: PMC8617492 DOI: 10.1073/pnas.2114326118] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Down syndrome (DS) neurocognitive disabilities associated with trisomy 21 are known; however, gene changes within individual brain cells occurring with age are unknown. Here, we interrogated >170,000 cells from 29 aging DS and control brains using single-nucleus RNA sequencing. We observed increases in inhibitory-over-excitatory neurons, microglial activation in the youngest DS brains coinciding with overexpression of genes associated with microglial-mediated synaptic pruning, and overexpression of the chromosome 21 gene RUNX1 that may be a potential driving factor in microglial activation. Single-nucleus long-read sequencing revealed hundreds of thousands of unannotated RNA transcripts. These included diverse species for the Alzheimer’s disease gene—amyloid precursor protein—that contained intra-exonic junctions previously associated with somatic gene recombination, which was also identified in ∼8,000 other genes. Down syndrome (DS), trisomy of human chromosome 21 (HSA21), is characterized by lifelong cognitive impairments and the development of the neuropathological hallmarks of Alzheimer’s disease (AD). The cellular and molecular modifications responsible for these effects are not understood. Here we performed single-nucleus RNA sequencing (snRNA-seq) employing both short- (Illumina) and long-read (Pacific Biosciences) sequencing technologies on a total of 29 DS and non-DS control prefrontal cortex samples. In DS, the ratio of inhibitory-to-excitatory neurons was significantly increased, which was not observed in previous reports examining sporadic AD. DS microglial transcriptomes displayed AD-related aging and activation signatures in advance of AD neuropathology, with increased microglial expression of C1q complement genes (associated with dendritic pruning) and the HSA21 transcription factor gene RUNX1. Long-read sequencing detected vast RNA isoform diversity within and among specific cell types, including numerous sequences that differed between DS and control brains. Notably, over 8,000 genes produced RNAs containing intra-exonic junctions, including amyloid precursor protein (APP) that had previously been associated with somatic gene recombination. These and related results illuminate large-scale cellular and transcriptomic alterations as features of the aging DS brain.
Collapse
|
10
|
De Cristofaro R, Sanguinetti M. Vaccine-induced thrombotic thrombocytopenia, a rare but severe case of friendly fire in the battle against COVID-19 pandemic: What pathogenesis? Eur J Intern Med 2021; 91:88-89. [PMID: 34244023 PMCID: PMC8238658 DOI: 10.1016/j.ejim.2021.06.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/08/2021] [Accepted: 06/17/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Raimondo De Cristofaro
- Dipartimento di Diagnostica per immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario "Gemelli" IRCCS,and Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica S. Cuore, Facoltà di Medicina e Chirurgia "Agostino Gemelli", Rome, Italy.
| | - Maurizio Sanguinetti
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS and Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
11
|
Takeuchi H, Yamaga S, Sasaki N, Kuboniwa M, Matsusaki M, Amano A. Porphyromonas gingivalis induces penetration of lipopolysaccharide and peptidoglycan through the gingival epithelium via degradation of coxsackievirus and adenovirus receptor. Cell Microbiol 2021; 23:e13388. [PMID: 34448537 DOI: 10.1111/cmi.13388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/13/2021] [Accepted: 08/22/2021] [Indexed: 11/28/2022]
Abstract
Porphyromonas gingivalis is a major pathogen of human periodontitis and dysregulates innate immunity at the gingival epithelial surface. We previously reported that the bacterium specifically degrades junctional adhesion molecule 1 (JAM1), causing gingival epithelial barrier breakdown. However, the functions of other JAM family protein(s) in epithelial barrier dysregulation caused by P. gingivalis are not fully understood. The present results show that gingipains, Arg-specific or Lys-specific cysteine proteases produced by P. gingivalis, specifically degrade coxsackievirus and adenovirus receptor (CXADR), a JAM family protein, at R145 and K235 in gingival epithelial cells. In contrast, a gingipain-deficient P. gingivalis strain was found to be impaired in regard to degradation of CXADR. Furthermore, knockdown of CXADR in artificial gingival epithelium increased permeability to dextran 40 kDa, lipopolysaccharide and peptidoglycan, whereas overexpression of CXADR in a gingival epithelial tissue model prevented penetration by those agents following P. gingivalis infection. Together, these results suggest that P. gingivalis gingipains breach the stratified squamous epithelium barrier by degrading CXADR as well as JAM1, which allows for efficient transfer of bacterial virulence factors into subepithelial tissues. TAKEAWAYS: P. gingivalis, a periodontal pathogen, degraded coxsackievirus and adenovirus receptor (CXADR), a JAM family protein, in gingival epithelial tissues. P. gingivalis gingipains, cysteine proteases, degraded CXADR at R145 and K235. CXADR degradation by P. gingivalis caused increased permeability to lipopolysaccharide and peptidoglycan through gingival epithelial tissues.
Collapse
Affiliation(s)
- Hiroki Takeuchi
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Shunsuke Yamaga
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Naoko Sasaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Masae Kuboniwa
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan.,Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Atsuo Amano
- Department of Preventive Dentistry, Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
12
|
Hirai T, Sato A, Koizumi N, Kurioka Y, Suzuki Y, Kano J, Yamakawa M, Nomura T, Fujii M, Sakurai F, Mizuguchi H, Watanabe Y, Utoguchi N. The infectivity of progeny adenovirus in the presence of neutralizing antibody. J Gen Virol 2021; 102. [PMID: 33843575 PMCID: PMC8290266 DOI: 10.1099/jgv.0.001590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human adenoviruses (Ads), common pathogens that cause upper respiratory and gastrointestinal infections, are blocked by neutralizing antibodies (nAbs). However, Ads are not fully eliminated even in hosts with nAbs. In this study, we assessed the infectivity of progeny Ad serotype 5 (Ad5) in the presence of nAb. The infectivity of Ad5 was evaluated according to the expression of the Ad genome and reporter gene. Infection by wild-type Ad5 and Ad5 vector continued to increase until 3 days after infection even in the presence of nAb. We established an assay for determining the infection levels of progeny Ad5 using a sorting system with magnetic beads and observed little difference in progeny Ad5 counts in the presence and absence of nAb 1 day after infection. Moreover, progeny Ad5 in the presence of nAb more effectively infected coxsackievirus and adenovirus receptor (CAR)-positive cells than CAR-negative cells. We investigated the function of fiber proteins, which are the binding partners of CAR, during secondary infection, observing that fibre proteins spread from infected cells to adjacent cells in a CAR-dependent manner. In conclusion, this study revealed that progeny Ad5 could infect cells even in the presence of nAb, differing from the common features of the Ad5 infection cycle. Our findings may be useful for developing new therapeutic agents against Ad infection.
Collapse
Affiliation(s)
- Takamasa Hirai
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Anna Sato
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
- Cosmetic Science Laboratory, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Hyougo, Japan
| | - Naoya Koizumi
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
| | - Yoh Kurioka
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
| | - Yui Suzuki
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
| | - Junpei Kano
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
| | - Makie Yamakawa
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
| | - Tetsuya Nomura
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
| | - Makiko Fujii
- Laboratory of Physical Chemistry, School of Pharmacy, Nihon University, Chiba, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- iPS Cell-Based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Global Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka, Japan
| | - Yoshiteru Watanabe
- Department of Pharmacy, Tohoku Medical and Pharmaceutical University Hospital, Miyagi, Japan
| | - Naoki Utoguchi
- Department of Pharmaceutics and Biopharmaceutics, Showa Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
13
|
Shafraz O, Xie B, Yamada S, Sivasankar S. Mapping transmembrane binding partners for E-cadherin ectodomains. Proc Natl Acad Sci U S A 2020; 117:31157-31165. [PMID: 33229577 PMCID: PMC7733791 DOI: 10.1073/pnas.2010209117] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We combine proximity labeling and single molecule binding assays to discover transmembrane protein interactions in cells. We first screen for candidate binding partners by tagging the extracellular and cytoplasmic regions of a "bait" protein with BioID biotin ligase and identify proximal proteins that are biotin tagged on both their extracellular and intracellular regions. We then test direct binding interactions between proximal proteins and the bait, using single molecule atomic force microscope binding assays. Using this approach, we identify binding partners for the extracellular region of E-cadherin, an essential cell-cell adhesion protein. We show that the desmosomal proteins desmoglein-2 and desmocollin-3, the focal adhesion protein integrin-α2β1, the receptor tyrosine kinase ligand ephrin-B1, and the classical cadherin P-cadherin, all directly interact with E-cadherin ectodomains. Our data shows that combining extracellular and cytoplasmic proximal tagging with a biophysical binding assay increases the precision with which transmembrane ectodomain interactors can be identified.
Collapse
Affiliation(s)
- Omer Shafraz
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Bin Xie
- Biophysics Graduate Group, University of California, Davis, CA 95616
| | - Soichiro Yamada
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Sanjeevi Sivasankar
- Department of Biomedical Engineering, University of California, Davis, CA 95616;
- Biophysics Graduate Group, University of California, Davis, CA 95616
| |
Collapse
|
14
|
Weng CF, Huang CJ, Wu MH, Lee HHC, Ling TY. Co-Expression of Coxsackievirus/Adenovirus Receptors and Desmoglein 2 in Lung Adenocarcinoma: A Comprehensive Analysis of Bioinformatics and Tissue Microarrays. J Clin Med 2020; 9:jcm9113693. [PMID: 33217893 PMCID: PMC7698609 DOI: 10.3390/jcm9113693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/06/2020] [Accepted: 11/16/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction: Coxsackievirus/adenovirus receptors (CARs) and desmoglein-2 (DSG2) are similar molecules to adenovirus-based vectors in the cell membrane. They have been found to be associated with lung epithelial cell tumorigenesis and can be useful markers in predicting survival outcome in lung adenocarcinoma (LUAD). Methods: A gene ontology enrichment analysis disclosed that DSG2 was highly correlated with CAR. Survival analysis was then performed on 262 samples from the Cancer Genome Atlas, forming “Stage 1A” or “Stage 1B”. We therefore analyzed a tissue microarray (TMA) comprised of 108 lung samples and an immunohistochemical assay. Computer counting software was used to calculate the H-score of the immune intensity. Cox regression and Kaplan–Meier analyses were used to determine the prognostic value. Results: CAR and DSG2 genes are highly co-expressed in early stage LUAD and associated with significantly poorer survival (p = 0.0046). TMA also showed that CAR/DSG2 expressions were altered in lung cancer tissue. CAR in the TMA was correlated with proliferation, apoptosis, and epithelial–mesenchymal transition (EMT), while DSG2 was associated with proliferation only. The Kaplan–Meier survival analysis revealed that CAR, DSG2, or a co-expression of CAR/DSG2 was associated with poorer overall survival. Conclusions: The co-expression of CAR/DSG2 predicted a worse overall survival in LUAD. CAR combined with DSG2 expression can predict prognosis.
Collapse
Affiliation(s)
- Ching-Fu Weng
- Division of Pulmonary Medicine, Department of Internal Medicine, Hsinchu Cathay General Hospital, Hsinchu 300, Taiwan;
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 100, Taiwan
| | - Chi-Jung Huang
- Medical Research Center, Cathay General Hospital, Taipei 106, Taiwan;
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
| | - Mei-Hsuan Wu
- Teaching and Research Center, Hsinchu Cathay General Hospital, Hsinchu 300, Taiwan;
| | - Henry Hsin-Chung Lee
- School of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
- Department of Surgery, Hsinchu Cathay General Hospital, Hsinchu 300, Taiwan
- Graduate Institute of Translational and Interdisciplinary Medicine, College of Health Sciences and Technology, National Central University, Taoyuan 320, Taiwan
- Correspondence: (H.H.-C.L.); (T.-Y.L.); Tel.: +886-3-527-8999 (ext. 61346) (H.H.-C.L.); +886-2-2312-3456 (ext. 88322) (T.-Y.L.)
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 100, Taiwan
- Correspondence: (H.H.-C.L.); (T.-Y.L.); Tel.: +886-3-527-8999 (ext. 61346) (H.H.-C.L.); +886-2-2312-3456 (ext. 88322) (T.-Y.L.)
| |
Collapse
|
15
|
Rathjen FG. The CAR group of Ig cell adhesion proteins–Regulators of gap junctions? Bioessays 2020; 42:e2000031. [DOI: 10.1002/bies.202000031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/01/2020] [Indexed: 12/29/2022]
|
16
|
Moscoso CG, Steer CJ. The Evolution of Gene Therapy in the Treatment of Metabolic Liver Diseases. Genes (Basel) 2020; 11:genes11080915. [PMID: 32785089 PMCID: PMC7463482 DOI: 10.3390/genes11080915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/02/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Monogenic metabolic disorders of hepatic origin number in the hundreds, and for many, liver transplantation remains the only cure. Liver-targeted gene therapy is an attractive treatment modality for many of these conditions, and there have been significant advances at both the preclinical and clinical stages. Viral vectors, including retroviruses, lentiviruses, adenovirus-based vectors, adeno-associated viruses and simian virus 40, have differing safety, efficacy and immunogenic profiles, and several of these have been used in clinical trials with variable success. In this review, we profile viral vectors and non-viral vectors, together with various payloads, including emerging therapies based on RNA, that are entering clinical trials. Genome editing technologies are explored, from earlier to more recent novel approaches that are more efficient, specific and safe in reaching their target sites. The various curative approaches for the multitude of monogenic hepatic metabolic disorders currently at the clinical development stage portend a favorable outlook for this class of genetic disorders.
Collapse
Affiliation(s)
- Carlos G. Moscoso
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Correspondence: (C.G.M.); (C.J.S.); Tel.: +1-612-625-8999 (C.G.M. & C.J.S.); Fax: +1-612-625-5620 (C.G.M. & C.J.S.)
| | - Clifford J. Steer
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Correspondence: (C.G.M.); (C.J.S.); Tel.: +1-612-625-8999 (C.G.M. & C.J.S.); Fax: +1-612-625-5620 (C.G.M. & C.J.S.)
| |
Collapse
|
17
|
Wehbi A, Kremer EJ, Dopeso-Reyes IG. Location of the Cell Adhesion Molecule "Coxsackievirus and Adenovirus Receptor" in the Adult Mouse Brain. Front Neuroanat 2020; 14:28. [PMID: 32581729 PMCID: PMC7287018 DOI: 10.3389/fnana.2020.00028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/08/2020] [Indexed: 12/30/2022] Open
Abstract
The coxsackievirus and adenovirus receptor (CAR) is a single-pass transmembrane cell adhesion molecule (CAM). CAR is expressed in numerous mammalian tissues including the brain, heart, lung, and testes. In epithelial cells, CAR functions are typical of the quintessential roles of numerous CAMs. However, in the brain the multiple roles of CAR are poorly understood. To better understand the physiological role of CAR in the adult brain, characterizing its location is a primordial step to advance our knowledge of its functions. In addition, CAR is responsible for the attachment, internalization, and retrograde transport of canine adenovirus type 2 (CAV-2) vectors, which have found a niche in the mapping of neuronal circuits and gene transfer to treat and model neurodegenerative diseases. In this study, we used immunohistochemistry and immunofluorescence to document the global location of CAR in the healthy, young adult mouse brain. Globally, we found that CAR is expressed by maturing and mature neurons in the brain parenchyma and located on the soma and on projections. While CAR occasionally colocalizes with glial fibrillary acidic protein, this overlap was restricted to areas that are associated with adult neurogenesis.
Collapse
Affiliation(s)
- Amani Wehbi
- Institut de Génétique Moléculaire de Montpellier, CNRS, Université de Montpellier, Montpellier, France
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS, Université de Montpellier, Montpellier, France
| | - Iria G Dopeso-Reyes
- Institut de Génétique Moléculaire de Montpellier, CNRS, Université de Montpellier, Montpellier, France
| |
Collapse
|
18
|
Albumin Enhances the Rate at Which Coxsackievirus B3 Strain 28 Converts to A-Particles. J Virol 2020; 94:JVI.01962-19. [PMID: 31915275 DOI: 10.1128/jvi.01962-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/21/2019] [Indexed: 12/13/2022] Open
Abstract
Three strains of coxsackievirus B3 (CVB3) differ by single mutations in capsid protein VP1 or VP3 and also differ in stability at 37°C in tissue culture medium. Among these strains, the CVB3/28 parent strain has been found to be uniquely sensitive to a component in fetal bovine serum (FBS) identified as serum albumin. In cell culture medium, serum increased the rate of CVB3/28 conversion to noninfectious particles at least 2-fold. The effect showed a saturable dose response. Rates of conversion to noninfectious virus with high concentrations of soluble coxsackievirus and adenovirus receptor (sCAR) were similar with and without FBS, but FBS amplified the catalytic effect of 100 nM sCAR nearly 3-fold. Such effects in other systems are due to nonessential activating cofactors.IMPORTANCE A factor other than the virus receptor expressed by target cells has been found to accelerate the loss of an enterovirus (CVB3/28) infectious titer, with little effect on nearly identical mutant strains. The destabilizing factor in fetal bovine serum, identified as albumin, does not interfere with the catalytic activity of soluble receptor at saturating receptor concentrations and amplifies the catalytic activity of the soluble receptor at a concentration that otherwise produces about one-third the saturated receptor-catalyzed rate of virus decay. This finding evidences the possibility that other virus-"priming" ligands may also be nonessential activating cofactors that serve to accelerate receptor-catalyzed viral eclipse.
Collapse
|
19
|
Heinemann U, Schuetz A. Structural Features of Tight-Junction Proteins. Int J Mol Sci 2019; 20:E6020. [PMID: 31795346 PMCID: PMC6928914 DOI: 10.3390/ijms20236020] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
Tight junctions are complex supramolecular entities composed of integral membrane proteins, membrane-associated and soluble cytoplasmic proteins engaging in an intricate and dynamic system of protein-protein interactions. Three-dimensional structures of several tight-junction proteins or their isolated domains have been determined by X-ray crystallography, nuclear magnetic resonance spectroscopy, and cryo-electron microscopy. These structures provide direct insight into molecular interactions that contribute to the formation, integrity, or function of tight junctions. In addition, the known experimental structures have allowed the modeling of ligand-binding events involving tight-junction proteins. Here, we review the published structures of tight-junction proteins. We show that these proteins are composed of a limited set of structural motifs and highlight common types of interactions between tight-junction proteins and their ligands involving these motifs.
Collapse
Affiliation(s)
- Udo Heinemann
- Macromolecular Structure and Interaction Laboratory, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Anja Schuetz
- Protein Production & Characterization Platform, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| |
Collapse
|
20
|
Li SJ, Vaughan A, Sturgill JF, Kepecs A. A Viral Receptor Complementation Strategy to Overcome CAV-2 Tropism for Efficient Retrograde Targeting of Neurons. Neuron 2019; 98:905-917.e5. [PMID: 29879392 DOI: 10.1016/j.neuron.2018.05.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/30/2018] [Accepted: 05/17/2018] [Indexed: 12/25/2022]
Abstract
Retrogradely transported neurotropic viruses enable genetic access to neurons based on their long-range projections and have become indispensable tools for linking neural connectivity with function. A major limitation of viral techniques is that they rely on cell-type-specific molecules for uptake and transport. Consequently, viruses fail to infect variable subsets of neurons depending on the complement of surface receptors expressed (viral tropism). We report a receptor complementation strategy to overcome this by potentiating neurons for the infection of the virus of interest-in this case, canine adenovirus type-2 (CAV-2). We designed AAV vectors for expressing the coxsackievirus and adenovirus receptor (CAR) throughout candidate projection neurons. CAR expression greatly increased retrograde-labeling rates, which we demonstrate for several long-range projections, including some resistant to other retrograde-labeling techniques. Our results demonstrate a receptor complementation strategy to abrogate endogenous viral tropism and thereby facilitate efficient retrograde targeting for functional analysis of neural circuits.
Collapse
Affiliation(s)
- Shu-Jing Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | | | - Adam Kepecs
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
21
|
Wrackmeyer U, Kaldrack J, Jüttner R, Pannasch U, Gimber N, Freiberg F, Purfürst B, Kainmueller D, Schmitz D, Haucke V, Rathjen FG, Gotthardt M. The cell adhesion protein CAR is a negative regulator of synaptic transmission. Sci Rep 2019; 9:6768. [PMID: 31043663 PMCID: PMC6494904 DOI: 10.1038/s41598-019-43150-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 04/17/2019] [Indexed: 11/09/2022] Open
Abstract
The Coxsackievirus and adenovirus receptor (CAR) is essential for normal electrical conductance in the heart, but its role in the postnatal brain is largely unknown. Using brain specific CAR knockout mice (KO), we discovered an unexpected role of CAR in neuronal communication. This includes increased basic synaptic transmission at hippocampal Schaffer collaterals, resistance to fatigue, and enhanced long-term potentiation. Spontaneous neurotransmitter release and speed of endocytosis are increased in KOs, accompanied by increased expression of the exocytosis associated calcium sensor synaptotagmin 2. Using proximity proteomics and binding studies, we link CAR to the exocytosis machinery as it associates with syntenin and synaptobrevin/VAMP2 at the synapse. Increased synaptic function does not cause adverse effects in KO mice, as behavior and learning are unaffected. Thus, unlike the connexin-dependent suppression of atrioventricular conduction in the cardiac knockout, communication in the CAR deficient brain is improved, suggesting a role for CAR in presynaptic processes.
Collapse
Affiliation(s)
- Uta Wrackmeyer
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Joanna Kaldrack
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - René Jüttner
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany.,Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Ulrike Pannasch
- Neuroscience Research Center, Cluster of Excellence NeuroCure, Charité, 10117, Berlin, Germany
| | - Niclas Gimber
- Department of Molecular Pharmacology and Cell Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Fabian Freiberg
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Bettina Purfürst
- Core Facility Electron Microscopy, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Dagmar Kainmueller
- Biomedical Image Analysis, Max Delbrück Center for Molecular Medicine and Berlin Institute of Health, 13125, Berlin, Germany
| | - Dietmar Schmitz
- Neuroscience Research Center, Cluster of Excellence NeuroCure, Charité, 10117, Berlin, Germany
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Fritz G Rathjen
- Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Michael Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany.
| |
Collapse
|
22
|
Abstract
Synapse formation is mediated by a surprisingly large number and wide variety of genes encoding many different protein classes. One of the families increasingly implicated in synapse wiring is the immunoglobulin superfamily (IgSF). IgSF molecules are by definition any protein containing at least one Ig-like domain, making this family one of the most common protein classes encoded by the genome. Here, we review the emerging roles for IgSF molecules in synapse formation specifically in the vertebrate brain, focusing on examples from three classes of IgSF members: ( a) cell adhesion molecules, ( b) signaling molecules, and ( c) immune molecules expressed in the brain. The critical roles for IgSF members in regulating synapse formation may explain their extensive involvement in neuropsychiatric and neurodevelopmental disorders. Solving the IgSF code for synapse formation may reveal multiple new targets for rescuing IgSF-mediated deficits in synapse formation and, eventually, new treatments for psychiatric disorders caused by altered IgSF-induced synapse wiring.
Collapse
Affiliation(s)
- Scott Cameron
- Center for Neuroscience, University of California, Davis, California 95618, USA; ,
| | | |
Collapse
|
23
|
Liem DA, Murali S, Sigdel D, Shi Y, Wang X, Shen J, Choi H, Caufield JH, Wang W, Ping P, Han J. Phrase mining of textual data to analyze extracellular matrix protein patterns across cardiovascular disease. Am J Physiol Heart Circ Physiol 2018; 315:H910-H924. [PMID: 29775406 DOI: 10.1152/ajpheart.00175.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Extracellular matrix (ECM) proteins have been shown to play important roles regulating multiple biological processes in an array of organ systems, including the cardiovascular system. Using a novel bioinformatics text-mining tool, we studied six categories of cardiovascular disease (CVD), namely, ischemic heart disease, cardiomyopathies, cerebrovascular accident, congenital heart disease, arrhythmias, and valve disease, anticipating novel ECM protein-disease and protein-protein relationships hidden within vast quantities of textual data. We conducted a phrase-mining analysis, delineating the relationships of 709 ECM proteins with the 6 groups of CVDs reported in 1,099,254 abstracts. The technology pipeline known as Context-Aware Semantic Online Analytical Processing was applied to semantically rank the association of proteins to each CVD and all six CVDs, performing analyses to quantify each protein-disease relationship. We performed principal component analysis and hierarchical clustering of the data, where each protein was visualized as a six-dimensional vector. We found that ECM proteins display variable degrees of association with the six CVDs; certain CVDs share groups of associated proteins, whereas others have divergent protein associations. We identified 82 ECM proteins sharing associations with all 6 CVDs. Our bioinformatics analysis ascribed distinct ECM pathways (via Reactome) from this subset of proteins, namely, insulin-like growth factor regulation and interleukin-4 and interleukin-13 signaling, suggesting their contribution to the pathogenesis of all six CVDs. Finally, we performed hierarchical clustering analysis and identified protein clusters predominantly associated with a targeted CVD; analyses of these proteins revealed unexpected insights underlying the key ECM-related molecular pathogenesis of each CVD, including virus assembly and release in arrhythmias. NEW & NOTEWORTHY The present study is the first application of a text-mining algorithm to characterize the relationships of 709 extracellular matrix-related proteins with 6 categories of cardiovascular disease described in 1,099,254 abstracts. Our analysis informed unexpected extracellular matrix functions, pathways, and molecular relationships implicated in the six cardiovascular diseases.
Collapse
Affiliation(s)
- David A Liem
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Sanjana Murali
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Dibakar Sigdel
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Yu Shi
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| | - Xuan Wang
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| | - Jiaming Shen
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| | - Howard Choi
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - John H Caufield
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Wei Wang
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Heart Big Data to Knowledge Center, Department of Computer Science, Scalable Analytics Institute, Henry Samueli School of Engineering and Applied Science, University of California , Los Angeles, California
| | - Peipei Ping
- NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Departments of Physiology, Medicine/Cardiology, and Bioinformatics, David Geffen School of Medicine, University of California , Los Angeles, California.,NIH BD2K Program Centers of Excellence for Big Data Computing-Heart BD2K Center, Heart Big Data to Knowledge Center, Department of Computer Science, Scalable Analytics Institute, Henry Samueli School of Engineering and Applied Science, University of California , Los Angeles, California
| | - JiaWei Han
- NIH BD2K Program Centers of Excellence for Big Data Computing-KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign , Champaign, Illinois
| |
Collapse
|
24
|
Coxsackievirus Adenovirus Receptor Loss Impairs Adult Neurogenesis, Synapse Content, and Hippocampus Plasticity. J Neurosci 2017; 36:9558-71. [PMID: 27629708 DOI: 10.1523/jneurosci.0132-16.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 07/26/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Although we are beginning to understand the late stage of neurodegenerative diseases, the molecular defects associated with the initiation of impaired cognition are poorly characterized. Here, we demonstrate that in the adult brain, the coxsackievirus and adenovirus receptor (CAR) is located on neuron projections, at the presynapse in mature neurons, and on the soma of immature neurons in the hippocampus. In a proinflammatory or diseased environment, CAR is lost from immature neurons in the hippocampus. Strikingly, in hippocampi of patients at early stages of late-onset Alzheimer's disease (AD), CAR levels are significantly reduced. Similarly, in triple-transgenic AD mice, CAR levels in hippocampi are low and further reduced after systemic inflammation. Genetic deletion of CAR from the mouse brain triggers deficits in adult neurogenesis and synapse homeostasis that lead to impaired hippocampal plasticity and cognitive deficits. We propose that post-translational CAR loss of function contributes to cognitive defects in healthy and diseased-primed brains. SIGNIFICANCE STATEMENT This study addressed the role of the coxsackievirus and adenovirus receptor (CAR), a single-pass cell adhesion molecule, in the adult brain. Our results demonstrate that CAR is expressed by mature neurons throughout the brain. In addition, we propose divergent roles for CAR in immature neurons, during neurogenesis, and at the mature synapse. Notably, CAR loss of function also affects hippocampal plasticity.
Collapse
|
25
|
Ortiz-Zapater E, Santis G, Parsons M. CAR: A key regulator of adhesion and inflammation. Int J Biochem Cell Biol 2017; 89:1-5. [PMID: 28545889 DOI: 10.1016/j.biocel.2017.05.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 05/17/2017] [Accepted: 05/20/2017] [Indexed: 10/19/2022]
Abstract
The coxsackie and adenovirus receptor (CAR) is a transmembrane receptor that plays a key role in controlling adhesion between adjacent epithelial cells. CAR is highly expressed in epithelial cells and was originally identified as a primary receptor for adenovirus cell binding. However, studies over the last 10 years have demonstrated that CAR plays a key role in co-ordinating cell-cell adhesion under homeostatic conditions including neuronal and cardiac development and cell junction stability; it has also been implicated in pathological states such as cancer growth and leukocyte transmigration during inflammation. Here we provide an overview of the functions of CAR as an adhesion molecule and highlight the emerging important role for CAR in controlling both recruitment of immune cells and in tumorigenesis.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, SE1 1UL, UK; Division of Asthma, Allergy & Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London, SE1 1UL, UK
| | - George Santis
- Division of Asthma, Allergy & Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London, SE1 1UL, UK
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London, SE1 1UL, UK.
| |
Collapse
|
26
|
Zussy C, Loustalot F, Junyent F, Kremer EJ, Salinas S. [The coxsackievirus and adenovirus receptor is involved in cognitive processes]. Med Sci (Paris) 2017; 33:89-92. [PMID: 28120763 DOI: 10.1051/medsci/20173301016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Charleine Zussy
- Institut de génétique moléculaire de Montpellier, CNRS 5535, 1919, route de Mende, 34293 Montpellier Cedex 5, France - Université de Montpellier, 4, boulevard Henry IV, 34967 Montpellier Cedex 2, France
| | - Fabien Loustalot
- Institut de génétique moléculaire de Montpellier, CNRS 5535, 1919, route de Mende, 34293 Montpellier Cedex 5, France - Université de Montpellier, 4, boulevard Henry IV, 34967 Montpellier Cedex 2, France
| | - Felix Junyent
- Institut de génétique moléculaire de Montpellier, CNRS 5535, 1919, route de Mende, 34293 Montpellier Cedex 5, France - Université de Montpellier, 4, boulevard Henry IV, 34967 Montpellier Cedex 2, France
| | - Eric J Kremer
- Institut de génétique moléculaire de Montpellier, CNRS 5535, 1919, route de Mende, 34293 Montpellier Cedex 5, France - Université de Montpellier, 4, boulevard Henry IV, 34967 Montpellier Cedex 2, France
| | - Sara Salinas
- UMR 1058, Inserm, Université de Montpellier, Établissement français du sang, Pathogenèse et contrôle des infections chroniques, 60, rue de Navacelles, 34394 Montpellier Cedex 5, France
| |
Collapse
|
27
|
Yap EH, Fiser A. ProtLID, a Residue-Based Pharmacophore Approach to Identify Cognate Protein Ligands in the Immunoglobulin Superfamily. Structure 2016; 24:2217-2226. [PMID: 27889206 PMCID: PMC5444293 DOI: 10.1016/j.str.2016.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 07/26/2016] [Accepted: 10/25/2016] [Indexed: 10/20/2022]
Abstract
Members of the extracellular immunoglobulin superfamily (IgSF) play a key role in immune regulation through the control of the co-stimulatory pathway, and have emerged as potent drug targets in cancers, infectious diseases, and autoimmunity. Despite the difficult experimental access to this class of proteins, single structures of ectodomains of IgSF proteins are solved with regularity. However, the most biologically critical challenge for this class of proteins is the identification of their cognate ligands that communicate intercellular signals. We describe a conceptually novel method, protein-ligand interface design (ProtLID), to identify cognate ligands from a subproteome for a given target receptor protein. ProtLID designs an optimal protein interface for a given receptor by running extensive molecular dynamics simulations of single-residue probes. The type and location of residue preferences establish a residue-based pharmacophore, which is subsequently used to find potential matches among candidate ligands within a subproteome.
Collapse
Affiliation(s)
- Eng-Hui Yap
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
28
|
Matthäus C, Langhorst H, Schütz L, Jüttner R, Rathjen FG. Cell-cell communication mediated by the CAR subgroup of immunoglobulin cell adhesion molecules in health and disease. Mol Cell Neurosci 2016; 81:32-40. [PMID: 27871939 DOI: 10.1016/j.mcn.2016.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022] Open
Abstract
The immunoglobulin superfamily represents a diverse set of cell-cell contact proteins and includes well-studied members such as NCAM1, DSCAM, L1 or the contactins which are strongly expressed in the nervous system. In this review we put our focus on the biological function of a less understood subgroup of Ig-like proteins composed of CAR (coxsackievirus and adenovirus receptor), CLMP (CAR-like membrane protein) and BT-IgSF (brain and testis specific immunoglobulin superfamily). The CAR-related proteins are type I transmembrane proteins containing an N-terminal variable (V-type) and a membrane proximal constant (C2-type) Ig domain in their extracellular region which are implicated in homotypic adhesion. They are highly expressed during embryonic development in a variety of tissues including the nervous system whereby in adult stages the protein level of CAR and CLMP decreases, only BT-IgSF expression increases within age. CAR-related proteins are concentrated at specialized cell-cell communication sites such as gap or tight junctions and are present at the plasma membrane in larger protein complexes. Considerable progress has been made on the molecular structure and interactions of CAR while research on CLMP and BT-IgSF is at an early stage. Studies on mouse mutants revealed biological functions of CAR in the heart and for CLMP in the gastrointestinal and urogenital systems. Furthermore, CAR and BT-IgSF appear to regulate synaptic function in the hippocampus.
Collapse
Affiliation(s)
- Claudia Matthäus
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany.
| | - Hanna Langhorst
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany
| | - Laura Schütz
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany
| | - René Jüttner
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany
| | - Fritz G Rathjen
- Max-Delbrück-Center for Molecular Medicine, Developmental Neurobiology, 13092 Berlin, Germany.
| |
Collapse
|
29
|
Gonzalez-Lozano MA, Klemmer P, Gebuis T, Hassan C, van Nierop P, van Kesteren RE, Smit AB, Li KW. Dynamics of the mouse brain cortical synaptic proteome during postnatal brain development. Sci Rep 2016; 6:35456. [PMID: 27748445 PMCID: PMC5066275 DOI: 10.1038/srep35456] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 09/28/2016] [Indexed: 01/04/2023] Open
Abstract
Development of the brain involves the formation and maturation of numerous synapses. This process requires prominent changes of the synaptic proteome and potentially involves thousands of different proteins at every synapse. To date the proteome analysis of synapse development has been studied sparsely. Here, we analyzed the cortical synaptic membrane proteome of juvenile postnatal days 9 (P9), P15, P21, P27, adolescent (P35) and different adult ages P70, P140 and P280 of C57Bl6/J mice. Using a quantitative proteomics workflow we quantified 1560 proteins of which 696 showed statistically significant differences over time. Synaptic proteins generally showed increased levels during maturation, whereas proteins involved in protein synthesis generally decreased in abundance. In several cases, proteins from a single functional molecular entity, e.g., subunits of the NMDA receptor, showed differences in their temporal regulation, which may reflect specific synaptic development features of connectivity, strength and plasticity. SNARE proteins, Snap 29/47 and Stx 7/8/12, showed higher expression in immature animals. Finally, we evaluated the function of Cxadr that showed high expression levels at P9 and a fast decline in expression during neuronal development. Knock down of the expression of Cxadr in cultured primary mouse neurons revealed a significant decrease in synapse density.
Collapse
Affiliation(s)
- Miguel A Gonzalez-Lozano
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Patricia Klemmer
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Titia Gebuis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Chopie Hassan
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Pim van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| |
Collapse
|
30
|
The Coxsackievirus and Adenovirus Receptor: Glycosylation and the Extracellular D2 Domain Are Not Required for Coxsackievirus B3 Infection. J Virol 2016; 90:5601-5610. [PMID: 27030267 DOI: 10.1128/jvi.00315-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/23/2016] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED The coxsackievirus and adenovirus receptor (CAR) is a member of the immunoglobulin superfamily (IgSF) and functions as a receptor for coxsackie B viruses (CVBs). The extracellular portion of CAR comprises two glycosylated immunoglobulin-like domains, D1 and D2. CAR-D1 binds to the virus and is essential for virus infection; however, it is not known whether D2 is also important for infection, and the role of glycosylation has not been explored. To understand the function of these structural components in CAR-mediated CVB3 infection, we generated a panel of human (h) CAR deletion and substitution mutants and analyzed their functionality as CVB receptors, examining both virus binding and replication. Lack of glycosylation of the CAR-D1 or -D2 domains did not adversely affect CVB3 binding or infection, indicating that the glycosylation of CAR is not required for its receptor functions. Deletion of the D2 domain reduced CVB3 binding, with a proportionate reduction in the efficiency of virus infection. Replacement of D2 with the homologous D2 domain from chicken CAR, or with the heterologous type C2 immunoglobulin-like domain from IgSF11, another IgSF member, fully restored receptor function; however, replacement of CAR-D2 with domains from CD155 or CD80 restored function only in part. These data indicate that glycosylation of the extracellular domain of hCAR plays no role in CVB3 receptor function and that CAR-D2 is not specifically required. The D2 domain may function largely as a spacer permitting virus access to D1; however, the data may also suggest that D2 affects virus binding by influencing the conformation of D1. IMPORTANCE An important step in virus infection is the initial interaction of the virus with its cellular receptor. Although the role in infection of the extracellular CAR-D1, cytoplasmic, and transmembrane domains have been analyzed extensively, nothing is known about the function of CAR-D2 and the extracellular glycosylation of CAR. Our data indicate that glycosylation of the extracellular CAR domain has only minor importance for the function of CAR as CVB3 receptor and that the D2 domain is not essential per se but contributes to receptor function by promoting the exposure of the D1 domain on the cell surface. These results contribute to our understanding of the coxsackievirus-receptor interactions.
Collapse
|
31
|
Murakami K, Eguchi J, Hida K, Nakatsuka A, Katayama A, Sakurai M, Choshi H, Furutani M, Ogawa D, Takei K, Otsuka F, Wada J. Antiobesity Action of ACAM by Modulating the Dynamics of Cell Adhesion and Actin Polymerization in Adipocytes. Diabetes 2016; 65:1255-67. [PMID: 26956488 DOI: 10.2337/db15-1304] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/22/2016] [Indexed: 11/13/2022]
Abstract
Coxsackie virus and adenovirus receptor-like membrane protein (CLMP) was identified as the tight junction-associated transmembrane protein of epithelial cells with homophilic binding activities. CLMP is also recognized as adipocyte adhesion molecule (ACAM), and it is upregulated in mature adipocytes in rodents and humans with obesity. Here, we present that aP2 promoter-driven ACAM transgenic mice are protected from obesity and diabetes with the prominent reduction of adipose tissue mass and smaller size of adipocytes. ACAM is abundantly expressed on plasma membrane of mature adipocytes and associated with formation of phalloidin-positive polymerized form of cortical actin (F-actin). By electron microscopy, the structure of zonula adherens with an intercellular space of ∼10-20 nm was observed with strict parallelism of the adjoining cell membranes over distances of 1-20 μm, where ACAM and γ-actin are abundantly expressed. The formation of zonula adherens may increase the mechanical strength, inhibit the adipocyte hypertrophy, and improve the insulin sensitivity.
Collapse
MESH Headings
- 3T3-L1 Cells
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/pathology
- Actin Cytoskeleton/ultrastructure
- Adherens Junctions/metabolism
- Adherens Junctions/pathology
- Adherens Junctions/ultrastructure
- Adipocytes, White/cytology
- Adipocytes, White/metabolism
- Adipocytes, White/pathology
- Adipocytes, White/ultrastructure
- Adiposity
- Animals
- Cell Adhesion
- Cell Size
- Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics
- Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism
- Diabetes Mellitus/etiology
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Diabetes Mellitus/prevention & control
- Diet, High-Fat/adverse effects
- Dietary Sucrose/adverse effects
- Female
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Microscopy, Immunoelectron
- Obesity/etiology
- Obesity/metabolism
- Obesity/pathology
- Obesity/prevention & control
- Recombinant Fusion Proteins/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Kazutoshi Murakami
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Eguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuyuki Hida
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Atsuko Nakatsuka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akihiro Katayama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Miwa Sakurai
- Department of Diabetic Nephropathy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Haruki Choshi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masumi Furutani
- Central Research Laboratory, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Daisuke Ogawa
- Department of Diabetic Nephropathy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kohji Takei
- Department of Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
32
|
Jang S, Oh D, Lee Y, Hosy E, Shin H, van Riesen C, Whitcomb D, Warburton JM, Jo J, Kim D, Kim SG, Um SM, Kwon SK, Kim MH, Roh JD, Woo J, Jun H, Lee D, Mah W, Kim H, Kaang BK, Cho K, Rhee JS, Choquet D, Kim E. Synaptic adhesion molecule IgSF11 regulates synaptic transmission and plasticity. Nat Neurosci 2016; 19:84-93. [PMID: 26595655 PMCID: PMC5010778 DOI: 10.1038/nn.4176] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022]
Abstract
Synaptic adhesion molecules regulate synapse development and plasticity through mechanisms that include trans-synaptic adhesion and recruitment of diverse synaptic proteins. We found that the immunoglobulin superfamily member 11 (IgSF11), a homophilic adhesion molecule that preferentially expressed in the brain, is a dual-binding partner of the postsynaptic scaffolding protein PSD-95 and AMPA glutamate receptors (AMPARs). IgSF11 required PSD-95 binding for its excitatory synaptic localization. In addition, IgSF11 stabilized synaptic AMPARs, as determined by IgSF11 knockdown-induced suppression of AMPAR-mediated synaptic transmission and increased surface mobility of AMPARs, measured by high-throughput, single-molecule tracking. IgSF11 deletion in mice led to the suppression of AMPAR-mediated synaptic transmission in the dentate gyrus and long-term potentiation in the CA1 region of the hippocampus. IgSF11 did not regulate the functional characteristics of AMPARs, including desensitization, deactivation or recovery. These results suggest that IgSF11 regulates excitatory synaptic transmission and plasticity through its tripartite interactions with PSD-95 and AMPARs.
Collapse
Affiliation(s)
- Seil Jang
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Daeyoung Oh
- Department of Biomedical Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
- Department of Psychiatry, CHA Bundang Medical Center, CHA
University, Seoul, Korea
| | - Yeunkum Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Eric Hosy
- University of Bordeaux, Interdisciplinary Institute for
Neuroscience, France; CNRS UMR 5297, F-33000 Bordeaux, France
| | - Hyewon Shin
- Department of Biomedical Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Christoph van Riesen
- Department of Molecular Neurobiology, Max Planck Institute of
Experimental Medicine, D-37075 Göttingen, Germany
| | - Daniel Whitcomb
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
- Centre for Synaptic Plasticity, University of Bristol, Whitson
street, Bristol, UK
| | - Julia M. Warburton
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
| | - Jihoon Jo
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
- Department of Biomedical Sciences, Chonnam National University
Medical School, Gwangju, South Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Sun Gyun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Seung Min Um
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Seok-kyu Kwon
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Myoung-Hwan Kim
- Department of Physiology, Seoul National University College of
Medicine, Seoul 110-799, Republic of Korea
- Seoul National University Bundang Hospital, Seongnam, Gyeonggi
463-707, Republic of Korea
| | - Junyeop Daniel Roh
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Jooyeon Woo
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Heejung Jun
- Brain and Cognitive Sciences, College of Natural Sciences, Seoul
National University, Seoul 151-747, Korea
| | - Dongmin Lee
- Department of Anatomy and Division of Brain Korea 21 Biomedical
Science, College of Medicine, Korea University, 126-1, 5-Ka, Anam-Dong, Seongbuk-Gu,
Seoul 136-705, Korea
| | - Won Mah
- Department of Anatomy and Neurobiology, School of Dentistry,
Kyungpook National University, Daegu 700-412, Korea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21 Biomedical
Science, College of Medicine, Korea University, 126-1, 5-Ka, Anam-Dong, Seongbuk-Gu,
Seoul 136-705, Korea
| | - Bong-Kiun Kaang
- Brain and Cognitive Sciences, College of Natural Sciences, Seoul
National University, Seoul 151-747, Korea
| | - Kwangwook Cho
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
- Centre for Synaptic Plasticity, University of Bristol, Whitson
street, Bristol, UK
| | - Jeong-Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of
Experimental Medicine, D-37075 Göttingen, Germany
| | - Daniel Choquet
- University of Bordeaux, Interdisciplinary Institute for
Neuroscience, France; CNRS UMR 5297, F-33000 Bordeaux, France
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| |
Collapse
|
33
|
Loustalot F, Kremer EJ, Salinas S. Membrane Dynamics and Signaling of the Coxsackievirus and Adenovirus Receptor. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 322:331-62. [PMID: 26940522 DOI: 10.1016/bs.ircmb.2015.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The coxsackievirus and adenovirus receptor (CAR) belongs to the immunoglobulin superfamily and acts as a receptor for some adenovirus types and group B coxsackieviruses. Its role is best described in epithelia where CAR participates to tight junction integrity and maintenance. Recently, several studies aimed to characterize its potential interaction with intracellular signaling pathways and highlighted several features linking CAR to gene expression. In addition, the molecular mechanisms leading to CAR-specific membrane targeting via the secretory pathway in polarized cells and its internalization are starting to be unraveled. This chapter discusses the interaction between membrane dynamics, intracellular trafficking, and signaling of CAR.
Collapse
Affiliation(s)
- Fabien Loustalot
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France.
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France.
| |
Collapse
|
34
|
Guo L, Tian S, Chen Y, Mao Y, Cui S, Hu A, Zhang J, Xia SL, Su Y, Du J, Block ER, Wang XL, Cui Z. CAT-1 as a novel CAM stabilizes endothelial integrity and mediates the protective actions of l-Arg via a NO-independent mechanism. J Mol Cell Cardiol 2015; 87:180-91. [DOI: 10.1016/j.yjmcc.2015.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 12/21/2022]
|
35
|
Levitt JA, Morton PE, Fruhwirth GO, Santis G, Chung PH, Parsons M, Suhling K. Simultaneous FRAP, FLIM and FAIM for measurements of protein mobility and interaction in living cells. BIOMEDICAL OPTICS EXPRESS 2015; 6:3842-54. [PMID: 26504635 PMCID: PMC4605044 DOI: 10.1364/boe.6.003842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/16/2015] [Accepted: 08/18/2015] [Indexed: 05/23/2023]
Abstract
We present a novel integrated multimodal fluorescence microscopy technique for simultaneous fluorescence recovery after photobleaching (FRAP), fluorescence lifetime imaging (FLIM) and fluorescence anisotropy imaging (FAIM). This approach captures a series of polarization-resolved fluorescence lifetime images during a FRAP recovery, maximizing the information available from a limited photon budget. We have applied this method to analyse the behaviour of GFP-labelled coxsackievirus and adenovirus receptor (CAR) in living human epithelial cells. Our data reveal that CAR exists in oligomeric states throughout the cell, and that these complexes occur in conjunction with high immobile fractions of the receptor at cell-cell junctions. These findings shed light on previously unknown molecular associations between CAR receptors in intact cells and demonstrate the power of combined FRAP, FLIM and FAIM microscopy as a robust method to analyse complex multi-component dynamics in living cells.
Collapse
Affiliation(s)
- James A. Levitt
- Department of Physics, King’s College London, Strand, London WC2R 2LS, UK
| | - Penny E. Morton
- Division of Asthma, Allergy, and Lung Biology, Guys Campus, King’s College London, London, UK
- Randall Division of Cell and Molecular Biophysics, Guys Campus, King’s College London, London, SE1 1UL, UK
| | - Gilbert O. Fruhwirth
- Department of Imaging Chemsitry and Biology, Division of Imaging Sciences and Biomedical Engineering, St. Thomas Hospital, King's College London, SE1 7EH, UK
| | - George Santis
- Division of Asthma, Allergy, and Lung Biology, Guys Campus, King’s College London, London, UK
| | - Pei-Hua Chung
- Department of Physics, King’s College London, Strand, London WC2R 2LS, UK
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, Guys Campus, King’s College London, London, SE1 1UL, UK
| | - Klaus Suhling
- Department of Physics, King’s College London, Strand, London WC2R 2LS, UK
| |
Collapse
|
36
|
Piersanti S, Burla R, Licursi V, Brito C, La Torre M, Alves PM, Simao D, Mottini C, Salinas S, Negri R, Tagliafico E, Kremer EJ, Saggio I. Transcriptional Response of Human Neurospheres to Helper-Dependent CAV-2 Vectors Involves the Modulation of DNA Damage Response, Microtubule and Centromere Gene Groups. PLoS One 2015. [PMID: 26207738 PMCID: PMC4514711 DOI: 10.1371/journal.pone.0133607] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Brain gene transfer using viral vectors will likely become a therapeutic option for several disorders. Helper-dependent (HD) canine adenovirus type 2 vectors (CAV-2) are well suited for this goal. These vectors are poorly immunogenic, efficiently transduce neurons, are retrogradely transported to afferent structures in the brain and lead to long-term transgene expression. CAV-2 vectors are being exploited to unravel behavior, cognition, neural networks, axonal transport and therapy for orphan diseases. With the goal of better understanding and characterizing HD-CAV-2 for brain therapy, we analyzed the transcriptomic modulation induced by HD-CAV-2 in human differentiated neurospheres derived from midbrain progenitors. This 3D model system mimics several aspects of the dynamic nature of human brain. We found that differentiated neurospheres are readily transduced by HD-CAV-2 and that transduction generates two main transcriptional responses: a DNA damage response and alteration of centromeric and microtubule probes. Future investigations on the biochemistry of processes highlighted by probe modulations will help defining the implication of HD-CAV-2 and CAR receptor binding in enchaining these functional pathways. We suggest here that the modulation of DNA damage genes is related to viral DNA, while the alteration of centromeric and microtubule probes is possibly enchained by the interaction of the HD-CAV-2 fibre with CAR.
Collapse
Affiliation(s)
- Stefania Piersanti
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Romina Burla
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Mattia La Torre
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Daniel Simao
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Carla Mottini
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Rodolfo Negri
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
| | - Enrico Tagliafico
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Isabella Saggio
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
- Institute of Molecular Biology and Pathology, CNR, Rome, Italy
- * E-mail:
| |
Collapse
|
37
|
Christianson MG, Lo DC. Differential roles of Aβ processing in hypoxia-induced axonal damage. Neurobiol Dis 2015; 77:94-105. [PMID: 25771168 DOI: 10.1016/j.nbd.2015.02.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 01/28/2015] [Accepted: 02/11/2015] [Indexed: 11/25/2022] Open
Abstract
Axonopathy is a common and early phase in neurodegenerative and traumatic CNS diseases. Recent work suggests that amyloid β (Aβ) produced from amyloid precursor protein (APP) may be a critical downstream mediator of CNS axonopathy in CNS diseases, particularly those associated with hypoxia. We critically tested this hypothesis in an adult retinal explant system that preserves the three-dimensional organization of the retina while permitting direct imaging of two cardinal features of early-stage axonopathy: axonal structural integrity and axonal transport capacity. Using this system, we found via pharmacological inhibition and genetic deletion of APP that production of Aβ is a necessary step in structural compromise of retinal ganglion cell (RGC) axons induced by the disease-relevant stressor hypoxia. However, identical blockade of Aβ production was not sufficient to protect axons from associated hypoxia-induced reduction in axonal transport. Thus, Aβ mediates distinct facets of hypoxia-induced axonopathy and may represent a functionally selective pharmacological target for therapies directed against early-stage axonopathy in CNS diseases.
Collapse
Affiliation(s)
- Melissa G Christianson
- Center for Drug Discovery and Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | - Donald C Lo
- Center for Drug Discovery and Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
38
|
Hwang JY, Lee KM, Kim YH, Shim HM, Bae YK, Hwang JH, Park H. Pregnancy loss following coxsackievirus b3 infection in mice during early gestation due to high expression of coxsackievirus-adenovirus receptor (CAR) in uterus and embryo. Exp Anim 2014; 63:63-72. [PMID: 24521864 PMCID: PMC4160930 DOI: 10.1538/expanim.63.63] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Coxsackieviruses are important pathogens in children and the outcomes of neonatal
infection can be serious or fatal. However, the outcomes of coxsackievirus infection
during early gestation are not well defined. In this study, we examined the possibility of
vertical transmission of coxsackievirus B3 (CVB3) and the effects of CVB3 infection on
early pregnancy of ICR mice. We found that the coxsackievirus and adenovirus receptor
(CAR) was highly expressed not only in embryos but also in the uterus of ICR mice. CVB3
replicated in the uterus 1 to 7 days post-infection (dpi), with the highest titer at 3
dpi. The pregnancy loss rate in mice infected with CVB3 during early gestation was 38.3%,
compared to 4.7% and 2.7% in mock-infected and UV-inactivated-CVB3 infected pregnant mice,
respectively. These data suggest that the uterus and embryo, which express abundant CAR,
are important targets of CVB3 and that the vertical transmission of CVB3 during early
gestation induces pregnancy loss.
Collapse
Affiliation(s)
- Ji Young Hwang
- Department of Microbiology, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Namgu, Daegu 705-717, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
39
|
Weber DA, Sumagin R, McCall IC, Leoni G, Neumann PA, Andargachew R, Brazil JC, Medina-Contreras O, Denning TL, Nusrat A, Parkos CA. Neutrophil-derived JAML inhibits repair of intestinal epithelial injury during acute inflammation. Mucosal Immunol 2014; 7:1221-32. [PMID: 24621992 PMCID: PMC4340686 DOI: 10.1038/mi.2014.12] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/20/2014] [Accepted: 02/04/2014] [Indexed: 02/04/2023]
Abstract
Neutrophil transepithelial migration (TEM) during acute inflammation is associated with mucosal injury. Using models of acute mucosal injury in vitro and in vivo, we describe a new mechanism by which neutrophils infiltrating the intestinal mucosa disrupt epithelial homeostasis. We report that junctional adhesion molecule-like protein (JAML) is cleaved from neutrophil surface by zinc metalloproteases during TEM. Neutrophil-derived soluble JAML binds to the epithelial tight junction protein coxsackie-adenovirus receptor (CAR) resulting in compromised barrier and inhibition of wound repair, through decreased epithelial proliferation. The deleterious effects of JAML on barrier and wound repair are reversed with an anti-JAML monoclonal antibody that inhibits JAML-CAR binding. JAML released from transmigrating neutrophils across inflamed epithelia may thus promote recruitment of leukocytes and aid in clearance of invading microorganisms. However, sustained release of JAML under pathologic conditions associated with persistence of large numbers of infiltrated neutrophils would compromise intestinal barrier and inhibit mucosal healing. Thus, targeting JAML-CAR interactions may improve mucosal healing responses under conditions of dysregulated neutrophil recruitment.
Collapse
Affiliation(s)
- Dominique A. Weber
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Ronen Sumagin
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Ingrid C. McCall
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Giovanna Leoni
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Philipp A. Neumann
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Rakieb Andargachew
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Jennifer C. Brazil
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Oscar Medina-Contreras
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Timothy L. Denning
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine
| | - Charles A. Parkos
- Epithelial Pathobiology and Mucosal Inflammation Unit, Department of Pathology and Laboratory Medicine,Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
40
|
The Ig CAM CAR is Implicated in Cardiac Development and Modulates Electrical Conduction in the Mature Heart. J Cardiovasc Dev Dis 2014. [DOI: 10.3390/jcdd1010111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
41
|
Interspecies differences in virus uptake versus cardiac function of the coxsackievirus and adenovirus receptor. J Virol 2014; 88:7345-56. [PMID: 24741103 DOI: 10.1128/jvi.00104-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED The coxsackievirus and adenovirus receptor (CAR) is a cell contact protein with an important role in virus uptake. Its extracellular immunoglobulin domains mediate the binding to coxsackievirus and adenovirus as well as homophilic and heterophilic interactions between cells. The cytoplasmic tail links CAR to the cytoskeleton and intracellular signaling cascades. In the heart, CAR is crucial for embryonic development, electrophysiology, and coxsackievirus B infection. Noncardiac functions are less well understood, in part due to the lack of suitable animal models. Here, we generated a transgenic mouse that rescued the otherwise embryonic-lethal CAR knockout (KO) phenotype by expressing chicken CAR exclusively in the heart. Using this rescue model, we addressed interspecies differences in coxsackievirus uptake and noncardiac functions of CAR. Survival of the noncardiac CAR KO (ncKO) mouse indicates an essential role for CAR in the developing heart but not in other tissues. In adult animals, cardiac activity was normal, suggesting that chicken CAR can replace the physiological functions of mouse CAR in the cardiomyocyte. However, chicken CAR did not mediate virus entry in vivo, so that hearts expressing chicken instead of mouse CAR were protected from infection and myocarditis. Comparison of sequence homology and modeling of the D1 domain indicate differences between mammalian and chicken CAR that relate to the sites important for virus binding but not those involved in homodimerization. Thus, CAR-directed anticoxsackievirus therapy with only minor adverse effects in noncardiac tissue could be further improved by selectively targeting the virus-host interaction while maintaining cardiac function. IMPORTANCE Coxsackievirus B3 (CVB3) is one of the most common human pathogens causing myocarditis. Its receptor, the coxsackievirus and adenovirus receptor (CAR), not only mediates virus uptake but also relates to cytoskeletal organization and intracellular signaling. Animals without CAR die prenatally with major cardiac malformations. In the adult heart, CAR is important for virus entry and electrical conduction, but its nonmuscle functions are largely unknown. Here, we show that chicken CAR expression exclusively in the heart can rescue the otherwise embryonic-lethal CAR knockout phenotype but does not support CVB3 infection of adult cardiomyocytes. Our findings have implications for the evolution of virus-host versus physiological interactions involving CAR and could help to improve future coxsackievirus-directed therapies inhibiting virus replication while maintaining CAR's cellular functions.
Collapse
|
42
|
Bissel SJ, Winkler CC, DelTondo J, Wang G, Williams K, Wiley CA. Coxsackievirus B4 myocarditis and meningoencephalitis in newborn twins. Neuropathology 2014; 34:429-437. [PMID: 24702280 DOI: 10.1111/neup.12121] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/14/2014] [Accepted: 03/16/2014] [Indexed: 11/29/2022]
Abstract
Coxsackievirus B4 (CB4) is a picornavirus associated with a variety of human diseases, including neonatal meningoencephalitis, myocarditis and type 1 diabetes. We report the pathological findings in twin newborns who died during an acute infection. The twins were born 1 month premature but were well and neurologically intact at birth. After a week they developed acute lethal neonatal sepsis and seizures. Histopathology demonstrated meningoencephalitis and severe myocarditis, as well as pancreatitis, adrenal medullitis and nephritis. Abundant CB4 sequences were identified in nucleic acid extracted from the brain and heart. In situ hybridization with probes to CB4 demonstrated infection of neurons, myocardiocytes, endocrine pancreas and adrenal medulla. The distribution of infected cells and immune response is consistent with reported clinical symptomatology where systemic and neurological diseases are the result of CB4 infection of select target cells.
Collapse
Affiliation(s)
- Stephanie J Bissel
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Caitlin C Winkler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Joseph DelTondo
- Allegheny County Medical Examiner, Pittsburgh, Pennsylvania, USA
| | - Guoji Wang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Karl Williams
- Allegheny County Medical Examiner, Pittsburgh, Pennsylvania, USA
| | - Clayton A Wiley
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
43
|
Luissint AC, Nusrat A, Parkos CA. JAM-related proteins in mucosal homeostasis and inflammation. Semin Immunopathol 2014; 36:211-26. [PMID: 24667924 DOI: 10.1007/s00281-014-0421-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/25/2014] [Indexed: 02/06/2023]
Abstract
Mucosal surfaces are lined by epithelial cells that form a physical barrier protecting the body against external noxious substances and pathogens. At a molecular level, the mucosal barrier is regulated by tight junctions (TJs) that seal the paracellular space between adjacent epithelial cells. Transmembrane proteins within TJs include junctional adhesion molecules (JAMs) that belong to the cortical thymocyte marker for Xenopus family of proteins. JAM family encompasses three classical members (JAM-A, JAM-B, and JAM-C) and related molecules including JAM4, JAM-like protein, Coxsackie and adenovirus receptor (CAR), CAR-like membrane protein and endothelial cell-selective adhesion molecule. JAMs have multiple functions that include regulation of endothelial and epithelial paracellular permeability, leukocyte recruitment during inflammation, angiogenesis, cell migration, and proliferation. In this review, we summarize the current knowledge regarding the roles of the JAM family members in the regulation of mucosal homeostasis and leukocyte trafficking with a particular emphasis on barrier function and its perturbation during pathological inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Epithelial pathobiology and mucosal inflammation research unit, Department of Pathology and Laboratory Medicine, Emory University, 615 Michael Street, 30306, Atlanta, GA, USA
| | | | | |
Collapse
|
44
|
Kinetic and structural analysis of coxsackievirus B3 receptor interactions and formation of the A-particle. J Virol 2014; 88:5755-65. [PMID: 24623425 DOI: 10.1128/jvi.00299-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED The coxsackievirus and adenovirus receptor (CAR) has been identified as the cellular receptor for group B coxsackieviruses, including serotype 3 (CVB3). CAR mediates infection by binding to CVB3 and catalyzing conformational changes in the virus that result in formation of the altered, noninfectious A-particle. Kinetic analyses show that the apparent first-order rate constant for the inactivation of CVB3 by soluble CAR (sCAR) at physiological temperatures varies nonlinearly with sCAR concentration. Cryo-electron microscopy (cryo-EM) reconstruction of the CVB3-CAR complex resulted in a 9.0-Å resolution map that was interpreted with the four available crystal structures of CAR, providing a consensus footprint for the receptor binding site. The analysis of the cryo-EM structure identifies important virus-receptor interactions that are conserved across picornavirus species. These conserved interactions map to variable antigenic sites or structurally conserved regions, suggesting a combination of evolutionary mechanisms for receptor site preservation. The CAR-catalyzed A-particle structure was solved to a 6.6-Å resolution and shows significant rearrangement of internal features and symmetric interactions with the RNA genome. IMPORTANCE This report presents new information about receptor use by picornaviruses and highlights the importance of attaining at least an ∼9-Å resolution for the interpretation of cryo-EM complex maps. The analysis of receptor binding elucidates two complementary mechanisms for preservation of the low-affinity (initial) interaction of the receptor and defines the kinetics of receptor-catalyzed conformational change to the A-particle.
Collapse
|
45
|
Schreiber J, Langhorst H, Jüttner R, Rathjen FG. The IgCAMs CAR, BT-IgSF, and CLMP: Structure, Function, and Diseases. ADVANCES IN NEUROBIOLOGY 2014; 8:21-45. [DOI: 10.1007/978-1-4614-8090-7_2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Berezin V, Walmod PS, Filippov M, Dityatev A. Targeting of ECM molecules and their metabolizing enzymes and receptors for the treatment of CNS diseases. PROGRESS IN BRAIN RESEARCH 2014; 214:353-88. [DOI: 10.1016/b978-0-444-63486-3.00015-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
47
|
Abstract
Vectors derived from the canine adenovirus serotype 2 (CAV-2) possess a high neurotropism and efficient retrograde transport that lead to widespread neuronal transduction in the central nervous system (CNS) of various animals. These abilities are due to the engagement of virions to the coxsackievirus and adenovirus receptor at the surface of neurons, which is linked to the endocytic and axonal transport machineries. The trafficking of CAV-2 and the coxsackievirus and adenovirus receptor (CAR) can be visualized ex vivo by incubating primary neurons (e.g., motoneurons and hippocampal neurons) with fluorescently labeled virions or recombinant viral proteins. Using this approach, we could recapitulate the mechanisms responsible for long-range transport of adenovirus in neurons.
Collapse
Affiliation(s)
- Charleine Zussy
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Universités de Montpellier I & II, Montpellier, France
| | | |
Collapse
|
48
|
Salinas S, Zussy C, Loustalot F, Henaff D, Menendez G, Morton PE, Parsons M, Schiavo G, Kremer EJ. Disruption of the coxsackievirus and adenovirus receptor-homodimeric interaction triggers lipid microdomain- and dynamin-dependent endocytosis and lysosomal targeting. J Biol Chem 2013; 289:680-95. [PMID: 24273169 DOI: 10.1074/jbc.m113.518365] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The coxsackievirus and adenovirus receptor (CAR) serves as a docking factor for some adenovirus (AdV) types and group B coxsackieviruses. Its role in AdV internalization is unclear as studies suggest that its intracellular domain is dispensable for some AdV infection. We previously showed that in motor neurons, AdV induced CAR internalization and co-transport in axons, suggesting that CAR was linked to endocytic and long-range transport machineries. Here, we characterized the mechanisms of CAR endocytosis in neurons and neuronal cells. We found that CAR internalization was lipid microdomain-, actin-, and dynamin-dependent, and subsequently followed by CAR degradation in lysosomes. Moreover, ligands that disrupted the homodimeric CAR interactions in its D1 domains triggered an internalization cascade involving sequences in its intracellular tail.
Collapse
Affiliation(s)
- Sara Salinas
- From the Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, Universités de Montpellier I & II, Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Houri N, Huang KC, Nalbantoglu J. The Coxsackievirus and Adenovirus Receptor (CAR) undergoes ectodomain shedding and regulated intramembrane proteolysis (RIP). PLoS One 2013; 8:e73296. [PMID: 24015300 PMCID: PMC3756012 DOI: 10.1371/journal.pone.0073296] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/19/2013] [Indexed: 11/18/2022] Open
Abstract
The Coxsackievirus and Adenovirus Receptor (CAR) is a cell adhesion molecule originally characterized as a virus receptor but subsequently shown to be involved in physiological processes such as neuronal and heart development, epithelial tight junction integrity, and tumour suppression. Proteolysis of cell adhesion molecules and a wide variety of other cell surface proteins serves as a mechanism for protein turnover and, in some cases, cell signaling. Metalloproteases such as A Disintegrin and Metalloprotease (ADAM) family members cleave cell surface receptors to release their substrates' ectodomains, while the presenilin/ɣ-secretase complex mediates regulated intramembrane proteolysis (RIP), releasing intracellular domain fragments from the plasma membrane. In the case of some substrates such as Notch and amyloid precursor protein (APP), the released intracellular domains enter the nucleus to modulate gene expression. We report that CAR ectodomain is constitutively shed from glioma cells and developing neurons, and is also shed when cells are treated with the phorbol ester phorbol 12-myristate 13-acetate (PMA) and the calcium ionophore ionomycin. We identified ADAM10 as a sheddase of CAR using assays involving shRNA knockdown and rescue, overexpression of wild-type ADAM10 and inhibition of ADAM10 activity by addition of its prodomain. In vitro peptide cleavage, mass spectrometry and mutagenesis revealed the amino acids M224 to L227 of CAR as the site of ADAM10-mediated ectodomain cleavage. CAR also undergoes RIP by the presenilin/γ-secretase complex, and the intracellular domain of CAR enters the nucleus. Ectodomain shedding is a prerequisite for RIP of CAR. Thus, CAR belongs to the increasing list of cell surface molecules that undergo ectodomain shedding and that are substrates for ɣ-secretase-mediated RIP.
Collapse
Affiliation(s)
- Nadia Houri
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Kuo-Cheng Huang
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Josephine Nalbantoglu
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
50
|
Volkmer H, Schreiber J, Rathjen FG. Regulation of adhesion by flexible ectodomains of IgCAMs. Neurochem Res 2012; 38:1092-9. [PMID: 23054071 DOI: 10.1007/s11064-012-0888-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/10/2012] [Indexed: 01/06/2023]
Abstract
To perform their diverse biological functions the adhesion activities of the cell adhesion molecules of the immunoglobulin superfamily (IgCAMs) might be regulated by local clustering, proteolytical shedding of their ectodomains or rapid recycling to and from the plasma membrane. Another form of regulation of adhesion might be obtained through flexible ectodomains of IgCAMs which adopt distinct conformations and which in turn modulate their adhesion activity. Here, we discuss variations in the conformation of the extracellular domains of CEACAM1 and CAR that might influence their binding and signaling activities. Furthermore, we concentrate on alternative splicing of single domains and short segments in the extracellular regions of L1 subfamily members that might affect the organization of the N-terminal located Ig-like domains. In particular, we discuss variations of the linker sequence between Ig-like domains 2 and 3 (D2 and D3) that is required for the horseshoe conformation.
Collapse
Affiliation(s)
- Hansjürgen Volkmer
- Naturwissenschaftliches und Medizinisches Institut an der Universität Tübingen, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | | | | |
Collapse
|