1
|
Kamakura S, Hayase J, Kohda A, Iwakiri Y, Chishiki K, Izaki T, Sumimoto H. TMEM25 is a Par3-binding protein that attenuates claudin assembly during tight junction development. EMBO Rep 2024; 25:144-167. [PMID: 38177906 PMCID: PMC10897455 DOI: 10.1038/s44319-023-00018-0] [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: 01/10/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 01/06/2024] Open
Abstract
The tight junction (TJ) in epithelial cells is formed by integral membrane proteins and cytoplasmic scaffolding proteins. The former contains the claudin family proteins with four transmembrane segments, while the latter includes Par3, a PDZ domain-containing adaptor that organizes TJ formation. Here we show the single membrane-spanning protein TMEM25 localizes to TJs in epithelial cells and binds to Par3 via a PDZ-mediated interaction with its C-terminal cytoplasmic tail. TJ development during epithelial cell polarization is accelerated by depletion of TMEM25, and delayed by overexpression of TMEM25 but not by that of a C-terminally deleted protein, indicating a regulatory role of TMEM25. TMEM25 associates via its N-terminal extracellular domain with claudin-1 and claudin-2 to suppress their cis- and trans-oligomerizations, both of which participate in TJ strand formation. Furthermore, Par3 attenuates TMEM25-claudin association via binding to TMEM25, implying its ability to affect claudin oligomerization. Thus, the TJ protein TMEM25 appears to negatively regulate claudin assembly in TJ formation, which regulation is modulated by its interaction with Par3.
Collapse
Affiliation(s)
- Sachiko Kamakura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Junya Hayase
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Akira Kohda
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yuko Iwakiri
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kanako Chishiki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tomoko Izaki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.
| |
Collapse
|
2
|
CZUBAK-PROWIZOR KAMILA, SWIATKOWSKA MARIA. Junctional adhesion molecule-A (JAM-A) in gynecological cancers: Current state of knowledge. BIOCELL 2023. [DOI: 10.32604/biocell.2023.025677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
3
|
Liu Y, Bertels S, Reischl M, Peravali R, Bastmeyer M, Popova AA, Levkin PA. Droplet Microarray Based Screening Identifies Proteins for Maintaining Pluripotency of hiPSCs. Adv Healthc Mater 2022; 11:e2200718. [PMID: 35799451 PMCID: PMC11468593 DOI: 10.1002/adhm.202200718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/10/2022] [Indexed: 01/27/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are crucial for disease modeling, drug discovery, and personalized medicine. Animal-derived materials hinderapplications of hiPSCs in medical fields. Thus, novel and well-defined substrate coatings capable of maintaining hiPSC pluripotency are important for advancing biomedical applications of hiPSCs. Here a miniaturized droplet microarray (DMA) platform to investigate 11 well-defined proteins, their 55 binary and 165 ternary combinations for their ability to maintainpluripotency of hiPSCs when applied as a surface coating, is used. Using this screening approach, ten protein group coatings are identified, which promote significantly higher NANOG expression of hiPSCs in comparison with Matrigel coating. With two of the identified coatings, long-term pluripotency maintenance of hiPSCs and subsequent differentiation into three germ layers are achieved. Compared with conventional high-throughput screening (HTS) in 96-well plates, the DMA platform uses only 83 µL of protein solution (0.83 µg total protein) and only ≈2.8 × 105 cells, decreasing the amount of proteins and cells ≈860 and 25-fold, respectively. The identified proteins will be essential for research and applications using hiPSCs, while the DMA platform demonstrates great potential for miniaturized HTS of scarce cells or expensive materials such as recombinant proteins.
Collapse
Affiliation(s)
- Yanxi Liu
- Institute of Biological and Chemical Systems – Functional Molecular SystemsKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Sarah Bertels
- Zoological InstituteCell‐ and NeurobiologyKarlsruhe Institute of TechnologyFritz‐Haber‐Weg 476131KarlsruheGermany
| | - Markus Reischl
- Institute for Automation and Applied InformaticsKarlsruhe Institute of TechnologyHermann‐von Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Ravindra Peravali
- Institute of Biological and Chemical Systems – Biological Information ProcessingKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Martin Bastmeyer
- Zoological InstituteCell‐ and NeurobiologyKarlsruhe Institute of TechnologyFritz‐Haber‐Weg 476131KarlsruheGermany
- Institute of Biological and Chemical Systems – Biological Information ProcessingKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Anna A. Popova
- Institute of Biological and Chemical Systems – Functional Molecular SystemsKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
| | - Pavel A. Levkin
- Institute of Biological and Chemical Systems – Functional Molecular SystemsKarlsruhe Institute of TechnologyHermann‐von‐Helmholtz‐Platz 176344Eggenstein‐LeopoldshafenGermany
- Institute of Organic ChemistryKarlsruhe Institute of TechnologyKaiserstraße 1276131KarlsruheGermany
| |
Collapse
|
4
|
Wang J, Chen X. Junctional Adhesion Molecules: Potential Proteins in Atherosclerosis. Front Cardiovasc Med 2022; 9:888818. [PMID: 35872908 PMCID: PMC9302484 DOI: 10.3389/fcvm.2022.888818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Junctional adhesion molecules (JAMs) are cell-cell adhesion molecules of the immunoglobulin superfamily and are involved in the regulation of diverse atherosclerosis-related processes such as endothelial barrier maintenance, leucocytes transendothelial migration, and angiogenesis. To combine and further broaden related results, this review concluded the recent progress in the roles of JAMs and predicted future studies of JAMs in the development of atherosclerosis.
Collapse
Affiliation(s)
- Junqi Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaoping Chen,
| |
Collapse
|
5
|
Li M, Huang Q, Lv X, Small HJ, Li C. Integrative omics analysis highlights the immunomodulatory effects of the parasitic dinoflagellate hhematodinium on crustacean hemocytes. FISH & SHELLFISH IMMUNOLOGY 2022; 125:35-47. [PMID: 35526798 DOI: 10.1016/j.fsi.2022.04.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/24/2022] [Accepted: 04/29/2022] [Indexed: 06/14/2023]
Abstract
Parasitic dinoflagellates in genus Hematodinium have caused substantial economic losses to multiple commercially valuable marine crustaceans around the world. Recent efforts to better understand the life cycle and biology of the parasite have improved our understanding of the disease ecology. However, studies on the host-parasite interaction, especially how Hematodinium parasites evade the host immune response are lacking. To address this shortfall, we used the comprehensive omics approaches (miRNA transcriptomics, iTRAQ-based proteomics) to get insights into the host-parasite interaction between hemocytes from Portunus trituberculatus and Hematodinium perezi in the present study. The parasitic dinoflagellate H. perezi remodeled the miRNome and proteome of hemocytes from challenged hosts, modulated the host immune response at both post-transcriptional and translational levels and caused post-transcriptional regulation to the host immune response. Multiple important cellular and humoral immune-related pathways (ex. Apoptosis, Endocytosis, ECM-receptor interaction, proPO activation pathway, Toll-like signaling pathway, Jak-STAT signaling pathway) were significantly affected by Hematodinium parasites. Through modulation of the host miRNome, the host immune responses of nodulation, proPO activation and antimicrobial peptides were significantly suppressed. Cellular homeostasis was imbalanced via post-transcriptional dysregulation of the phagosome and peroxisome pathways. Cellular structure and communication was seriously impacted by post-transcriptional downregulation of ECM-receptor interaction and focal adhesion pathways. In conclusion, H. perezi parasites could trigger striking changes in the miRNome and proteome of crustacean hemocytes, and this parasite exhibited multifaceted immunomodulatory effects and potential immune-suppressive mechanisms in crustacean hosts.
Collapse
Affiliation(s)
- Meng Li
- CAS Key Lab of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Marine Ecology and Environmental Science Laboratory, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Centre for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Qian Huang
- CAS Key Lab of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyang Lv
- CAS Key Lab of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hamish J Small
- Virginia Institute of Marine Science, William & Mary, P.O. Box 1346, Gloucester Point, VA, 23062, USA.
| | - Caiwen Li
- CAS Key Lab of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Marine Ecology and Environmental Science Laboratory, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; Centre for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Fan S, Smith MS, Keeney J, O’Leary MN, Nusrat A, Parkos CA. JAM-A signals through the Hippo pathway to regulate intestinal epithelial proliferation. iScience 2022; 25:104316. [PMID: 35602956 PMCID: PMC9114518 DOI: 10.1016/j.isci.2022.104316] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/18/2022] [Accepted: 04/22/2022] [Indexed: 01/25/2023] Open
Abstract
JAM-A is a tight-junction-associated protein that contributes to regulation of intestinal homeostasis. We report that JAM-A interacts with NF2 and LATS1, functioning as an initiator of the Hippo signaling pathway, well-known for regulation of proliferation. Consistent with these findings, we observed increased YAP activity in JAM-A-deficient intestinal epithelial cells (IEC). Furthermore, overexpression of a dimerization-deficient mutant, JAM-A-DL1, failed to initiate Hippo signaling, phenocopying JAM-A-deficient IEC, whereas overexpression of JAM-A-WT activated Hippo signaling and suppressed proliferation. Lastly, we identify EVI1, a transcription factor reported to promote cellular proliferation, as a contributor to the pro-proliferative phenotype in JAM-A-DL1 overexpressing IEC downstream of YAP. Collectively, our findings establish a new role for JAM-A as a cell-cell contact sensor, raising implications for understanding the contribution(s) of JAM-A to IEC proliferation in the mammalian epithelium.
Collapse
Affiliation(s)
- Shuling Fan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michelle Sydney Smith
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Justin Keeney
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Monique N. O’Leary
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Charles A. Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Rath D, Rapp V, Schwartz J, Winter S, Emschermann F, Arnold D, Rheinlaender J, Büttcher M, Strebl M, Braun MB, Altgelt K, Uribe ÁP, Schories C, Canjuga D, Schaeffeler E, Borst O, Schäffer TE, Langer H, Stehle T, Schwab M, Geisler T, Gawaz M, Chatterjee M. Homophilic Interaction Between Transmembrane-JAM-A and Soluble JAM-A Regulates Thrombo-Inflammation: Implications for Coronary Artery Disease. JACC Basic Transl Sci 2022; 7:445-461. [PMID: 35663628 PMCID: PMC9156439 DOI: 10.1016/j.jacbts.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 11/02/2022]
Abstract
Genetic predisposition through F11R-single-nucleotide variation (SNV) influences circulatory soluble junctional adhesion molecule-A (sJAM-A) levels in coronary artery disease (CAD) patients. Homozygous carriers of the minor alleles (F11R-SNVs rs2774276, rs790056) show enhanced levels of thrombo-inflammatory sJAM-A. Both F11R-SNVs and sJAM-A are associated with worse prognosis for recurrent myocardial infarction in CAD patients. Platelet surface-associated JAM-A correlate with platelet activation markers in CAD patients. Activated platelets shed transmembrane-JAM-A, generating proinflammatory sJAM-A and JAM-A-bearing microparticles. Platelet transmembrane-JAM-A and sJAM-A as homophilic interaction partners exaggerate thrombotic and thrombo-inflammatory platelet monocyte interactions. Therapeutic strategies interfering with this homophilic interface may regulate thrombotic and thrombo-inflammatory platelet response in cardiovascular pathologies where circulatory sJAM-A levels are elevated.
Collapse
Key Words
- ACM, all-cause mortality
- ACS, acute coronary syndrome
- ADP, adenosine diphosphate
- CAD, coronary artery disease
- CCS, chronic coronary syndrome
- CE, combined endpoint
- HC, homozygous carriers
- IS, ischemic stroke
- JAM-A
- JAM-A, junctional adhesion molecule-A
- MI, myocardial infarction
- SNV
- SNV, single-nucleotide variation
- TRAP, thrombin receptor activating peptide
- coronary artery disease
- platelet
- sJAM-A, soluble junctional adhesion molecule-A
- smJAM-A, soluble murine junctional adhesion molecule-A
- thrombo-inflammation
Collapse
Affiliation(s)
- Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Vera Rapp
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Jessica Schwartz
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Stefan Winter
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany
| | - Frederic Emschermann
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Daniel Arnold
- Institute of Applied Physics, University of Tübingen, Tübingen, Germany
| | | | - Manuela Büttcher
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Michael Strebl
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Michael B. Braun
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Konstanze Altgelt
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Álvaro Petersen Uribe
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Schories
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Denis Canjuga
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Elke Schaeffeler
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
- DFG Heisenberg Group Thrombocardiology, Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | | | - Harald Langer
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Tübingen, Germany
- Department of Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
An Unusual Aspartic Acid Cluster in the Reovirus Attachment Fiber σ1 Mediates Stability at Low pH and Preserves Trimeric Organization. J Virol 2022; 96:e0033122. [PMID: 35380459 DOI: 10.1128/jvi.00331-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The reovirus attachment protein σ1 mediates cell attachment and receptor binding and is thought to undergo conformational changes during viral disassembly. σ1 is a trimeric filamentous protein with an α-helical coiled-coil tail, a triple-β-spiral body, and a globular head. At the trimer interface, the head domain features an unusual and conserved aspartic acid cluster, which forms the only significant intratrimer interactions in the head and must be protonated to allow trimer formation. To define the role of pH on σ1 stability and conformation, we tested its domains over a wide range of pH values. We show that all domains of σ1 are remarkably thermostable, even at the low pH of the stomach. We determined the optimal pH for stability to be between pHs 5 and 6, a value close to the pH of the endosome and of the jejunum. The σ1 head is stable at acidic and neutral pH but detrimerizes at basic pH. When Asp345 in the aspartic acid cluster is mutated to asparagine (D345N), the σ1 head loses stability at low pH and is more prone to detrimerize. Although the D345N mutation does not affect σ1 binding affinity for the JAM-A receptor, the overall binding stoichiometry is reduced by one-third. The additional replacement of the neighboring His349 with alanine disrupts inner trimer surface interactions, leading to a less thermostable and monomeric σ1 D345N head that fails to bind the JAM-A receptor. When the body is expressed together with the head domain, the thermostability is restored and the stoichiometry of the binding to JAM-A receptor is preserved. Our results confirm a fundamental role of the aspartic acid cluster as a pH-dependent molecular switch controlling trimerization and enhancing thermostability of σ1, which represent essential requirements to accomplish reovirus infection and entry and might be common mechanisms among other enteric viruses. IMPORTANCE Enteric viruses withstand the highly acidic environment of the stomach during transmission, and many of them use low pH as a trigger for conformational changes associated with entry. For many nonenveloped viruses, the structural basis of these effects is not clear. We have investigated the stability of the reovirus attachment protein σ1 over a range of pHs and find it to be remarkably thermostable, especially at low pH. We identify a role for the aspartic acid cluster in maintaining σ1 thermostability, trimeric organization, and binding to JAM-A receptor especially at the gastric pH reovirus has to withstand while passing the stomach. The understanding of monomer-trimer dynamics within σ1 enhances our knowledge of reovirus entry and has implications for stability and transmission of other enteric viruses.
Collapse
|
9
|
Smith YE, Wang G, Flynn CL, Madden SF, MacEneaney O, Cruz RGB, Richards CE, Jahns H, Brennan M, Cremona M, Hennessy BT, Sheehan K, Casucci A, Sani FA, Hudson L, Fay J, Vellanki SH, O’Flaherty S, Devocelle M, Hill ADK, Brennan K, Sukumar S, Hopkins AM. Functional Antagonism of Junctional Adhesion Molecule-A (JAM-A), Overexpressed in Breast Ductal Carcinoma In Situ (DCIS), Reduces HER2-Positive Tumor Progression. Cancers (Basel) 2022; 14:cancers14051303. [PMID: 35267611 PMCID: PMC8909510 DOI: 10.3390/cancers14051303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Specific drug targets for breast ductal carcinoma in situ (DCIS) remain elusive, despite increasing disease prevalence and burden to healthcare services. Estrogen receptor (ER)-negative HER2-positive DCIS, associated with the poorest patient prognosis, is in particular need of novel therapeutic avenues. This report provides the first evidence that a cell surface protein called JAM-A is upregulated on human DCIS patient tissues and can be readily targeted by a novel JAM-A-binding peptide inhibitor in separate in vivo models of DCIS. The anti-tumor efficacy and lack of systemic toxicity of this lead inhibitor, coupled with early indications of potential signaling pathways implicated, support the value of future studies investigating JAM-A as a novel drug target in DCIS patients. Abstract Breast ductal carcinoma in situ (DCIS) is clinically challenging, featuring high diagnosis rates and few targeted therapies. Expression/signaling from junctional adhesion molecule-A (JAM-A) has been linked to poor prognosis in invasive breast cancers, but its role in DCIS is unknown. Since progression from DCIS to invasive cancer has been linked with overexpression of the human epidermal growth factor receptor-2 (HER2), and JAM-A regulates HER2 expression, we evaluated JAM-A as a therapeutic target in DCIS. JAM-A expression was immunohistochemically assessed in patient DCIS tissues. A novel JAM-A antagonist (JBS2) was designed and tested alone/in combination with the HER2 kinase inhibitor lapatinib, using SUM-225 cells in vitro and in vivo as validated DCIS models. Murine tumors were proteomically analyzed. JAM-A expression was moderate/high in 96% of DCIS patient tissues, versus 23% of normal adjacent tissues. JBS2 bound to recombinant JAM-A, inhibiting cell viability in SUM-225 cells and a primary DCIS culture in vitro and in a chick embryo xenograft model. JBS2 reduced tumor progression in in vivo models of SUM-225 cells engrafted into mammary fat pads or directly injected into the mammary ducts of NOD-SCID mice. Preliminary proteomic analysis revealed alterations in angiogenic and apoptotic pathways. High JAM-A expression in aggressive DCIS lesions and their sensitivity to treatment by a novel JAM-A antagonist support the viability of testing JAM-A as a novel therapeutic target in DCIS.
Collapse
Affiliation(s)
- Yvonne E. Smith
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Guannan Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (G.W.); (S.S.)
| | - Ciara L. Flynn
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Stephen F. Madden
- Data Science Centre, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland;
| | - Owen MacEneaney
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Rodrigo G. B. Cruz
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Cathy E. Richards
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Hanne Jahns
- School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland;
| | - Marian Brennan
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland;
| | - Mattia Cremona
- Department of Medical Oncology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (M.C.); (B.T.H.)
| | - Bryan T. Hennessy
- Department of Medical Oncology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (M.C.); (B.T.H.)
| | - Katherine Sheehan
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Alexander Casucci
- School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (A.C.); (F.A.S.)
| | - Faizah A. Sani
- School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (A.C.); (F.A.S.)
| | - Lance Hudson
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Joanna Fay
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Sri H. Vellanki
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Siobhan O’Flaherty
- Department of Chemistry, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (S.O.); (M.D.)
| | - Marc Devocelle
- Department of Chemistry, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (S.O.); (M.D.)
| | - Arnold D. K. Hill
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Kieran Brennan
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (G.W.); (S.S.)
| | - Ann M. Hopkins
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
- Correspondence: ; Tel.: +353-1-809-3858
| |
Collapse
|
10
|
A Transcriptional Link between HER2, JAM-A and FOXA1 in Breast Cancer. Cells 2022; 11:cells11040735. [PMID: 35203384 PMCID: PMC8870165 DOI: 10.3390/cells11040735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 01/03/2023] Open
Abstract
Overexpression of the human epidermal growth factor receptor-2 (HER2) is associated with aggressive disease in breast and certain other cancers. At a cellular level, the adhesion protein Junctional Adhesion Molecule-A (JAM-A) has been reported to regulate the expression of HER3 via a transcriptional pathway involving FOXA1. Since FOXA1 is also a suggested transcription factor for HER2, this study set out to determine if JAM-A regulates HER2 expression via a similar mechanism. An integrated tripartite approach was taken, involving cellular expression studies after targeted disruption of individual players in the putative pathway, in silico identification of relevant HER2 promoter regions and, finally, interrogation of cancer patient survival databases to deconstruct functionally important links between HER2, JAM-A and FOXA1 gene expression. The outcome of these investigations revealed a unidirectional pathway in which JAM-A expression transcriptionally regulates that of HER2 by influencing the binding of FOXA1 to a specific site in the HER2 gene promoter. Moreover, a correlation between JAM-A and HER2 gene expression was identified in 75% of a sample of 40 cancer types from The Cancer Genome Atlas, and coincident high mean mRNA expression of JAM-A, HER2 and FOXA1 was associated with poorer survival outcomes in HER2-positive (but not HER2-negative) patients with either breast or gastric tumors. These investigations provide the first evidence of a transcriptional pathway linking JAM-A, HER2 and FOXA1 in cancer settings, and support potential future pharmacological targeting of JAM-A as an upstream regulator of HER2.
Collapse
|
11
|
Mendoza C, Nagidi SH, Collett K, Mckell J, Mizrachi D. Calcium regulates the interplay between the tight junction and epithelial adherens junction at the plasma membrane. FEBS Lett 2022; 596:219-231. [PMID: 34882783 DOI: 10.1002/1873-3468.14252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/05/2021] [Accepted: 12/05/2021] [Indexed: 01/15/2023]
Abstract
The apical junctional complex (AJC) is a membrane protein ultrastructure that regulates cell adhesion and homeostasis. The tight junction (TJ) and the adherens junction (AJ) are substructures of the AJC. The interplay between TJ and AJ membrane proteins to assemble the AJC remains unclear. We employed synthetic biology strategies to express the basic membrane elements of a simple AJC-the adhesive extracellular domains of junctional adhesion molecule A (JAM-A), epithelial cadherin, claudin 1, and occludin-to study their interactions. Our results suggest that calcium concentration fluctuations and JAM-A, acting as an interface molecule between the TJ and AJ, orchestrate their interplay. Calcium affects the secondary structure, oligomerization, and binding affinity of homotypic and heterotypic interactions of TJ and AJ components, thus acting as a molecular switch influencing AJC dynamics.
Collapse
Affiliation(s)
- Christopher Mendoza
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| | - Sai Harsha Nagidi
- Department of Molecular Microbiology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| | - Kjetil Collett
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| | - Jacob Mckell
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| | - Dario Mizrachi
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| |
Collapse
|
12
|
Tugizov S. Virus-associated disruption of mucosal epithelial tight junctions and its role in viral transmission and spread. Tissue Barriers 2021; 9:1943274. [PMID: 34241579 DOI: 10.1080/21688370.2021.19432749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023] Open
Abstract
Oropharyngeal, airway, intestinal, and genital mucosal epithelia are the main portals of entry for the majority of human pathogenic viruses. To initiate systemic infection, viruses must first be transmitted across the mucosal epithelium and then spread across the body. However, mucosal epithelia have well-developed tight junctions, which have a strong barrier function that plays a critical role in preventing the spread and dissemination of viral pathogens. Viruses can overcome these barriers by disrupting the tight junctions of mucosal epithelia, which facilitate paracellular viral penetration and initiate systemic disease. Disruption of tight and adherens junctions may also release the sequestered viral receptors within the junctional areas, and liberation of hidden receptors may facilitate viral infection of mucosal epithelia. This review focuses on possible molecular mechanisms of virus-associated disruption of mucosal epithelial junctions and its role in transmucosal viral transmission and spread.
Collapse
Affiliation(s)
- Sharof Tugizov
- Department of Medicine, School of Medicine, University of California-San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Tugizov S. Virus-associated disruption of mucosal epithelial tight junctions and its role in viral transmission and spread. Tissue Barriers 2021; 9:1943274. [PMID: 34241579 DOI: 10.1080/21688370.2021.1943274] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Oropharyngeal, airway, intestinal, and genital mucosal epithelia are the main portals of entry for the majority of human pathogenic viruses. To initiate systemic infection, viruses must first be transmitted across the mucosal epithelium and then spread across the body. However, mucosal epithelia have well-developed tight junctions, which have a strong barrier function that plays a critical role in preventing the spread and dissemination of viral pathogens. Viruses can overcome these barriers by disrupting the tight junctions of mucosal epithelia, which facilitate paracellular viral penetration and initiate systemic disease. Disruption of tight and adherens junctions may also release the sequestered viral receptors within the junctional areas, and liberation of hidden receptors may facilitate viral infection of mucosal epithelia. This review focuses on possible molecular mechanisms of virus-associated disruption of mucosal epithelial junctions and its role in transmucosal viral transmission and spread.
Collapse
Affiliation(s)
- Sharof Tugizov
- Department of Medicine, School of Medicine, University of California-San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Czubak-Prowizor K, Babinska A, Swiatkowska M. The F11 Receptor (F11R)/Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A) in cancer progression. Mol Cell Biochem 2021; 477:79-98. [PMID: 34533648 PMCID: PMC8755661 DOI: 10.1007/s11010-021-04259-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022]
Abstract
The F11 Receptor (F11R), also called Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A), is a transmembrane glycoprotein of the immunoglobulin superfamily, which is mainly located in epithelial and endothelial cell tight junctions and also expressed on circulating platelets and leukocytes. It participates in the regulation of various biological processes, as diverse as paracellular permeability, tight junction formation and maintenance, leukocyte transendothelial migration, epithelial-to-mesenchymal transition, angiogenesis, reovirus binding, and platelet activation. Dysregulation of F11R/JAM-A may result in pathological consequences and disorders in normal cell function. A growing body of evidence points to its role in carcinogenesis and invasiveness, but its tissue-specific pro- or anti-tumorigenic role remains a debated issue. The following review focuses on the F11R/JAM-A tissue-dependent manner in tumorigenesis and metastasis and also discusses the correlation between poor patient clinical outcomes and its aberrant expression. In the future, it will be required to clarify the signaling pathways that are activated or suppressed via the F11R/JAM-A protein in various cancer types to understand its multiple roles in cancer progression and further use it as a novel direct target for cancer treatment.
Collapse
Affiliation(s)
- Kamila Czubak-Prowizor
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland.
| | - Anna Babinska
- Department of Medicine, State University of New York Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY, 11203, USA
| | - Maria Swiatkowska
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland
| |
Collapse
|
15
|
Taylor A, Warner M, Mendoza C, Memmott C, LeCheminant T, Bailey S, Christensen C, Keller J, Suli A, Mizrachi D. Chimeric Claudins: A New Tool to Study Tight Junction Structure and Function. Int J Mol Sci 2021; 22:ijms22094947. [PMID: 34066630 PMCID: PMC8124314 DOI: 10.3390/ijms22094947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
The tight junction (TJ) is a structure composed of multiple proteins, both cytosolic and membranal, responsible for cell–cell adhesion in polarized endothelium and epithelium. The TJ is intimately connected to the cytoskeleton and plays a role in development and homeostasis. Among the TJ’s membrane proteins, claudins (CLDNs) are key to establishing blood–tissue barriers that protect organismal physiology. Recently, several crystal structures have been reported for detergent extracted recombinant CLDNs. These structural advances lack direct evidence to support quaternary structure of CLDNs. In this article, we have employed protein-engineering principles to create detergent-independent chimeric CLDNs, a combination of a 4-helix bundle soluble monomeric protein (PDB ID: 2jua) and the apical—50% of human CLDN1, the extracellular domain that is responsible for cell–cell adhesion. Maltose-binding protein-fused chimeric CLDNs (MBP-CCs) used in this study are soluble proteins that retain structural and functional aspects of native CLDNs. Here, we report the biophysical characterization of the structure and function of MBP-CCs. MBP-fused epithelial cadherin (MBP-eCAD) is used as a control and point of comparison of a well-characterized cell-adhesion molecule. Our synthetic strategy may benefit other families of 4-α-helix membrane proteins, including tetraspanins, connexins, pannexins, innexins, and more.
Collapse
|
16
|
Mendoza C, Nagidi SH, Mizrachi D. Molecular Characterization of the Extracellular Domain of Human Junctional Adhesion Proteins. Int J Mol Sci 2021; 22:ijms22073482. [PMID: 33801758 PMCID: PMC8037251 DOI: 10.3390/ijms22073482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/22/2022] Open
Abstract
The junction adhesion molecule (JAM) family of proteins play central roles in the tight junction (TJ) structure and function. In contrast to claudins (CLDN) and occludin (OCLN), the other membrane proteins of the TJ, whose structure is that of a 4α-helix bundle, JAMs are members of the immunoglobulin superfamily. The JAM family is composed of four members: A, B, C and 4. The crystal structure of the extracellular domain of JAM-A continues to be used as a template to model the secondary and tertiary structure of the other members of the family. In this article, we have expressed the extracellular domains of JAMs fused with maltose-binding protein (MBP). This strategy enabled the work presented here, since JAM-B, JAM-C and JAM4 are more difficult targets due to their more hydrophobic nature. Our results indicate that each member of the JAM family has a unique tertiary structure in spite of having similar secondary structures. Surface plasmon resonance (SPR) revealed that heterotypic interactions among JAM family members can be greatly favored compared to homotypic interactions. We employ the well characterized epithelial cadherin (E-CAD) as a means to evaluate the adhesive properties of JAMs. We present strong evidence that suggests that homotypic or heterotypic interactions among JAMs are stronger than that of E-CADs.
Collapse
Affiliation(s)
- Christopher Mendoza
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
| | - Sai Harsha Nagidi
- Department of Molecular Microbiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
| | - Dario Mizrachi
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
- Correspondence:
| |
Collapse
|
17
|
Bonilha CS, Benson RA, Brewer JM, Garside P. Targeting Opposing Immunological Roles of the Junctional Adhesion Molecule-A in Autoimmunity and Cancer. Front Immunol 2020; 11:602094. [PMID: 33324419 PMCID: PMC7723963 DOI: 10.3389/fimmu.2020.602094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/02/2020] [Indexed: 01/04/2023] Open
Abstract
The junctional adhesion molecule-A (JAM-A) is a cell surface adhesion molecule expressed on platelets, epithelial cells, endothelial cells and leukocytes (e. g. monocytes and dendritic cells). JAM-A plays a relevant role in leukocyte trafficking and its therapeutic potential has been studied in several pathological conditions due to its capacity to induce leukocyte migration out of inflamed sites or infiltration into tumor sites. However, disruption of JAM-A pathways may worsen clinical pathology in some cases. As such, the effects of JAM-A manipulation on modulating immune responses in the context of different diseases must be better understood. In this mini-review, we discuss the potential of JAM-A as a therapeutic target, summarizing findings from studies manipulating JAM-A in the context of inflammatory diseases (e.g. autoimmune diseases) and cancer and highlighting described mechanisms.
Collapse
Affiliation(s)
- Caio S. Bonilha
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Robert A. Benson
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
- Research and Development Department, Antibody Analytics Ltd., Newhouse, Lanarkshire, United Kingdom
| | - James M. Brewer
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Paul Garside
- College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
18
|
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: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/01/2020] [Indexed: 12/29/2022]
|
19
|
Generation of Genetically RGD σ1-Modified Oncolytic Reovirus That Enhances JAM-A-Independent Infection of Tumor Cells. J Virol 2020; 94:JVI.01703-20. [PMID: 32907973 DOI: 10.1128/jvi.01703-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/14/2022] Open
Abstract
Mammalian reovirus (MRV) strain type 3 Dearing (T3D) is a naturally occurring oncolytic virus that has been developed as a potential cancer therapeutic. However, MRV treatment cannot be applied to cancer cells expressing low levels of junctional adhesion molecule A (JAM-A), which is the entry receptor of MRV. In this study, we developed a reverse genetics system for MRV strain T3D-L, which showed high oncolytic potency. To modify the cell tropism of MRV, an arginine-glycine-aspartic acid (RGD) peptide with an affinity to integrin was inserted at the C terminus or loop structures of the viral cell attachment protein σ1. The recombinant RGD σ1-modified viruses induced remarkable cell lysis in human cancer cell lines with marginal JAM-A expression and in JAM-A knockout cancer cell lines generated by a CRISPR/Cas9 system. Pretreatment of cells with anti-integrin antibody decreased cell death caused by the RGD σ1-modified virus, suggesting the infection to the cells was via a specific interaction with integrin αV. By using mouse models, we assessed virulence of the RGD σ1-modified viruses in vivo This system will open new avenues for the use of genetically modified oncolytic MRV for use as a cancer therapy.IMPORTANCE Oncolytic viruses kill tumors without affecting normal cells. A variety of oncolytic viruses are used as cancer therapeutics. Mammalian reovirus (MRV), which belongs to the genus Orthoreovirus, family Reoviridae, is one such natural oncolytic virus. The anticancer effects of MRV are being evaluated in clinical trials. Unlike other oncolytic viruses, MRV has not been genetically modified for use as a cancer therapeutic in clinical trials. Here, we used a reverse genetic approach to introduce an integrin-affinity peptide sequence into the MRV cell attachment protein σ1 to alter the natural tropism of the virus. The recombinant viruses were able to infect cancer cell lines expressing very low levels of the MRV entry receptor, junctional adhesion molecule A (JAM-A), and cause tumor cell death while maintaining its original tropism via JAM-A. This is a novel report of a genetically modified oncolytic MRV by introducing a peptide sequence into σ1.
Collapse
|
20
|
Li M, Wang J, Huang Q, Li C. Proteomic analysis highlights the immune responses of the hepatopancreas against Hematodinium infection in Portunus trituberculatus. J Proteomics 2019; 197:92-105. [DOI: 10.1016/j.jprot.2018.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/15/2018] [Accepted: 11/16/2018] [Indexed: 12/20/2022]
|
21
|
Smith HQ, Smith TJ. The Dynamic Capsid Structures of the Noroviruses. Viruses 2019; 11:v11030235. [PMID: 30857192 PMCID: PMC6466125 DOI: 10.3390/v11030235] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 12/21/2022] Open
Abstract
Noroviruses are responsible for almost a fifth of all cases of gastroenteritis worldwide. New strains evolve every 2–4 years by escaping herd immunity and cause worldwide epidemics. In the US alone, noroviruses are responsible for ~20 million cases and more than 70,000 hospitalizations of infected children, annually. Efforts towards a vaccine have been hindered by a lack of detailed structural information about antibody binding and the mechanisms of antibody escape. Caliciviruses have 180 copies of the major capsid protein (VP1; ~58 kDa), that is divided into the N-terminus (N), the shell (S) and C-terminal protruding (P) domains. The S domain forms a shell around the viral RNA genome, while the P domains dimerize to form protrusions on the capsid surface. The P domain is subdivided into P1 and P2 subdomains, with the latter containing the binding sites for cellular receptors and neutralizing antibodies. There is increasing evidence that these viruses are extremely dynamic and this flexibility is critical for viral replication. There are at least two modes of flexibility; the entire P domain relative to the shell and within the P domain itself. Here, the details and possible roles for this remarkable flexibility will be reviewed.
Collapse
Affiliation(s)
- Hong Q Smith
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, TX 77555-0645, USA.
| | - Thomas J Smith
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, TX 77555-0645, USA.
| |
Collapse
|
22
|
Conley MJ, McElwee M, Azmi L, Gabrielsen M, Byron O, Goodfellow IG, Bhella D. Calicivirus VP2 forms a portal-like assembly following receptor engagement. Nature 2019; 565:377-381. [PMID: 30626974 DOI: 10.1038/s41586-018-0852-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/10/2018] [Indexed: 11/08/2022]
Abstract
To initiate infection, many viruses enter their host cells by triggering endocytosis following receptor engagement. However, the mechanisms by which non-enveloped viruses escape the endosome are poorly understood. Here we present near-atomic-resolution cryo-electron microscopy structures for feline calicivirus both undecorated and labelled with a soluble fragment of its cellular receptor, feline junctional adhesion molecule A. We show that VP2, a minor capsid protein encoded by all caliciviruses1,2, forms a large portal-like assembly at a unique three-fold axis of symmetry, following receptor engagement. This assembly-which was not detected in undecorated virions-is formed of twelve copies of VP2, arranged with their hydrophobic N termini pointing away from the virion surface. Local rearrangement at the portal site leads to the opening of a pore in the capsid shell. We hypothesize that the portal-like assembly functions as a channel for the delivery of the calicivirus genome, through the endosomal membrane, into the cytoplasm of a host cell, thereby initiating infection. VP2 was previously known to be critical for the production of infectious virus3; our findings provide insights into its structure and function that advance our understanding of the Caliciviridae.
Collapse
Affiliation(s)
- Michaela J Conley
- Medical Research Council University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marion McElwee
- Medical Research Council University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Liyana Azmi
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | | - Olwyn Byron
- School of Life Sciences, University of Glasgow, Glasgow, UK
| | - Ian G Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - David Bhella
- Medical Research Council University of Glasgow Centre for Virus Research, Glasgow, UK.
| |
Collapse
|
23
|
Abstract
Sialic acid-based glycoconjugates cover the surfaces of many different cell types, defining key properties of the cell surface such as overall charge or likely interaction partners. Because of this prominence, sialic acids play prominent roles in mediating attachment and entry to viruses belonging to many different families. In this review, we first describe how interactions between viruses and sialic acid-based glycan structures can be identified and characterized using a range of techniques. We then highlight interactions between sialic acids and virus capsid proteins in four different viruses, and discuss what these interactions have taught us about sialic acid engagement and opportunities to interfere with binding.
Collapse
Affiliation(s)
- Bärbel S Blaum
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
24
|
Fahey JK, Williams SM, Tyagi S, Powell DR, Hallab JC, Chahal G, Ramialison MS, White AJ. The Intercellular Tight Junction and Spontaneous Coronary Artery Dissection. J Am Coll Cardiol 2018; 72:1752-1753. [DOI: 10.1016/j.jacc.2018.07.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/04/2018] [Accepted: 07/02/2018] [Indexed: 10/28/2022]
|
25
|
Yeung J, Li W, Holinstat M. Platelet Signaling and Disease: Targeted Therapy for Thrombosis and Other Related Diseases. Pharmacol Rev 2018; 70:526-548. [PMID: 29925522 PMCID: PMC6013590 DOI: 10.1124/pr.117.014530] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Platelets are essential for clotting in the blood and maintenance of normal hemostasis. Under pathologic conditions such as atherosclerosis, vascular injury often results in hyperactive platelet activation, resulting in occlusive thrombus formation, myocardial infarction, and stroke. Recent work in the field has elucidated a number of platelet functions unique from that of maintaining hemostasis, including regulation of tumor growth and metastasis, inflammation, infection, and immune response. Traditional therapeutic targets for inhibiting platelet activation have primarily been limited to cyclooxygenase-1, integrin αIIbβ3, and the P2Y12 receptor. Recently identified signaling pathways regulating platelet function have made it possible to develop novel approaches for pharmacological intervention in the blood to limit platelet reactivity. In this review, we cover the newly discovered roles for platelets as well as their role in hemostasis and thrombosis. These new roles for platelets lend importance to the development of new therapies targeted to the platelet. Additionally, we highlight the promising receptor and enzymatic targets that may further decrease platelet activation and help to address the myriad of pathologic conditions now known to involve platelets without significant effects on hemostasis.
Collapse
Affiliation(s)
- Jennifer Yeung
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| | - Wenjie Li
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| | - Michael Holinstat
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
26
|
Lasswitz L, Chandra N, Arnberg N, Gerold G. Glycomics and Proteomics Approaches to Investigate Early Adenovirus-Host Cell Interactions. J Mol Biol 2018; 430:1863-1882. [PMID: 29746851 PMCID: PMC7094377 DOI: 10.1016/j.jmb.2018.04.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/24/2018] [Accepted: 04/30/2018] [Indexed: 12/14/2022]
Abstract
Adenoviruses as most viruses rely on glycan and protein interactions to attach to and enter susceptible host cells. The Adenoviridae family comprises more than 80 human types and they differ in their attachment factor and receptor usage, which likely contributes to the diverse tropism of the different types. In the past years, methods to systematically identify glycan and protein interactions have advanced. In particular sensitivity, speed and coverage of mass spectrometric analyses allow for high-throughput identification of glycans and peptides separated by liquid chromatography. Also, developments in glycan microarray technologies have led to targeted, high-throughput screening and identification of glycan-based receptors. The mapping of cell surface interactions of the diverse adenovirus types has implications for cell, tissue, and species tropism as well as drug development. Here we review known adenovirus interactions with glycan- and protein-based receptors, as well as glycomics and proteomics strategies to identify yet elusive virus receptors and attachment factors. We finally discuss challenges, bottlenecks, and future research directions in the field of non-enveloped virus entry into host cells.
Collapse
Affiliation(s)
- Lisa Lasswitz
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
| | - Naresh Chandra
- Department of Clinical Microbiology, Virology, Umeå University, SE-90185 Umeå, Sweden; Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90185 Umea, Sweden
| | - Niklas Arnberg
- Department of Clinical Microbiology, Virology, Umeå University, SE-90185 Umeå, Sweden; Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90185 Umea, Sweden.
| | - Gisa Gerold
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany; Department of Clinical Microbiology, Virology, Umeå University, SE-90185 Umeå, Sweden; Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umea, Sweden.
| |
Collapse
|
27
|
Maginnis MS. Virus-Receptor Interactions: The Key to Cellular Invasion. J Mol Biol 2018; 430:2590-2611. [PMID: 29924965 PMCID: PMC6083867 DOI: 10.1016/j.jmb.2018.06.024] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 06/04/2018] [Accepted: 06/08/2018] [Indexed: 11/05/2022]
Abstract
Virus–receptor interactions play a key regulatory role in viral host range, tissue tropism, and viral pathogenesis. Viruses utilize elegant strategies to attach to one or multiple receptors, overcome the plasma membrane barrier, enter, and access the necessary host cell machinery. The viral attachment protein can be viewed as the “key” that unlocks host cells by interacting with the “lock”—the receptor—on the cell surface, and these lock-and-key interactions are critical for viruses to successfully invade host cells. Many common themes have emerged in virus–receptor utilization within and across virus families demonstrating that viruses often target particular classes of molecules in order to mediate these events. Common viral receptors include sialylated glycans, cell adhesion molecules such as immunoglobulin superfamily members and integrins, and phosphatidylserine receptors. The redundancy in receptor usage suggests that viruses target particular receptors or “common locks” to take advantage of their cellular function and also suggests evolutionary conservation. Due to the importance of initial virus interactions with host cells in viral pathogenesis and the redundancy in viral receptor usage, exploitation of these strategies would be an attractive target for new antiviral therapeutics. Viral receptors are key regulators of host range, tissue tropism, and viral pathogenesis. Many viruses utilize common viral receptors including sialic acid, cell adhesion molecules such as immunoglobulin superfamily members and integrins, and phosphatidylserine receptors. Detailed molecular interactions between viruses and receptors have been defined through elegant biochemical analyses including glycan array screens, structural–functional analyses, and cell-based approaches providing tremendous insights into these initial events in viral infection. Commonalities in virus–receptor interactions present promising targets for the development of broad-spectrum antiviral therapies.
Collapse
Affiliation(s)
- Melissa S Maginnis
- Department of Molecular and Biomedical Sciences, The University of Maine, Orono, ME 04469-5735, USA.
| |
Collapse
|
28
|
Ma J, Fan Y, Zhou Y, Liu W, Jiang N, Zhang J, Zeng L. Efficient resistance to grass carp reovirus infection in JAM-A knockout cells using CRISPR/Cas9. FISH & SHELLFISH IMMUNOLOGY 2018; 76:206-215. [PMID: 29477498 DOI: 10.1016/j.fsi.2018.02.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 06/08/2023]
Abstract
The hemorrhagic disease of grass carp (Ctenopharyngodon idellus) induced by grass carp reovirus (GCRV) leads to huge economic losses in China and currently, there are no effective methods available for prevention and treatment. The various GCRV genotypes may be one of the major obstacles in the pursuit of an effective antiviral treatment. In this study, we exploited CRISPR/Cas9 gene editing to specifically knockout the DNA sequence of the grass carp Junctional Adhesion Molecule-A (gcJAM-A) and evaluated in vitro resistance against various GCRV genotypes. Our results show that CRISPR/Cas9 effectively knocked out gcJAM-A and reduced GCRV infection for two different genotypes in permissive grass carp kidney cells (CIK), as evidenced by suppressed cytopathic effect (CPE) and GCRV progeny production in infected cells. In addition, with ectopic expression of gcJAM-A in cells, non-permissive cells derived from Chinese giant salamander (Andrias davidianus) muscle (GSM) could be highly infected by both GCRV-JX0901 and Hubei grass carp disease reovirus (HGDRV) strains that have different genotypes. Taken together, the results demonstrate that gcJAM-A is necessary for GCRV infection, implying a potential approach for viral control in aquaculture.
Collapse
Affiliation(s)
- Jie Ma
- Division of Fish Disease, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei, 430223, PR China.
| | - Yuding Fan
- Division of Fish Disease, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei, 430223, PR China.
| | - Yong Zhou
- Division of Fish Disease, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei, 430223, PR China.
| | - Wenzhi Liu
- Division of Fish Disease, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei, 430223, PR China.
| | - Nan Jiang
- Division of Fish Disease, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei, 430223, PR China.
| | - Jieming Zhang
- Division of Fish Disease, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei, 430223, PR China.
| | - Lingbing Zeng
- Division of Fish Disease, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, Hubei, 430223, PR China.
| |
Collapse
|
29
|
Steinbacher T, Kummer D, Ebnet K. Junctional adhesion molecule-A: functional diversity through molecular promiscuity. Cell Mol Life Sci 2018; 75:1393-1409. [PMID: 29238845 PMCID: PMC11105642 DOI: 10.1007/s00018-017-2729-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 12/27/2022]
Abstract
Cell adhesion molecules (CAMs) of the immunoglobulin superfamily (IgSF) regulate important processes such as cell proliferation, differentiation and morphogenesis. This activity is primarily due to their ability to initiate intracellular signaling cascades at cell-cell contact sites. Junctional adhesion molecule-A (JAM-A) is an IgSF-CAM with a short cytoplasmic tail that has no catalytic activity. Nevertheless, JAM-A is involved in a variety of biological processes. The functional diversity of JAM-A resides to a large part in a C-terminal PDZ domain binding motif which directly interacts with nine different PDZ domain-containing proteins. The molecular promiscuity of its PDZ domain motif allows JAM-A to recruit protein scaffolds to specific sites of cell-cell adhesion and to assemble signaling complexes at those sites. Here, we review the molecular characteristics of JAM-A, including its dimerization, its interaction with scaffolding proteins, and the phosphorylation of its cytoplasmic domain, and we describe how these characteristics translate into diverse biological activities.
Collapse
Affiliation(s)
- Tim Steinbacher
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Daniel Kummer
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany.
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany.
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany.
| |
Collapse
|
30
|
Sutherland DM, Aravamudhan P, Dermody TS. An Orchestra of Reovirus Receptors: Still Searching for the Conductor. Adv Virus Res 2017; 100:223-246. [PMID: 29551138 DOI: 10.1016/bs.aivir.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Viruses are constantly engaged in a molecular arms race with the host, where efficient and tactical use of cellular receptors benefits critical steps in infection. Receptor use dictates initiation, establishment, and spread of viral infection to new tissues and hosts. Mammalian orthoreoviruses (reoviruses) are pervasive pathogens that use multiple receptors to overcome protective host barriers to disseminate from sites of initial infection and cause disease in young mammals. In particular, reovirus invades the central nervous system (CNS) with serotype-dependent tropism and disease. A single viral gene, encoding the attachment protein σ1, segregates with distinct patterns of CNS injury. Despite the identification and characterization of several reovirus receptors, host factors that dictate tropism via interaction with σ1 remain undefined. Here, we summarize the state of the reovirus receptor field and discuss open questions toward understanding how the reovirus attachment protein dictates CNS tropism.
Collapse
Affiliation(s)
| | | | - Terence S Dermody
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
31
|
Ebnet K. Junctional Adhesion Molecules (JAMs): Cell Adhesion Receptors With Pleiotropic Functions in Cell Physiology and Development. Physiol Rev 2017; 97:1529-1554. [PMID: 28931565 DOI: 10.1152/physrev.00004.2017] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
Junctional adhesion molecules (JAM)-A, -B and -C are cell-cell adhesion molecules of the immunoglobulin superfamily which are expressed by a variety of tissues, both during development and in the adult organism. Through their extracellular domains, they interact with other adhesion receptors on opposing cells. Through their cytoplasmic domains, they interact with PDZ domain-containing scaffolding and signaling proteins. In combination, these two properties regulate the assembly of signaling complexes at specific sites of cell-cell adhesion. The multitude of molecular interactions has enabled JAMs to adopt distinct cellular functions such as the regulation of cell-cell contact formation, cell migration, or mitotic spindle orientation. Not surprisingly, JAMs regulate diverse processes such as epithelial and endothelial barrier formation, hemostasis, angiogenesis, hematopoiesis, germ cell development, and the development of the central and peripheral nervous system. This review summarizes the recent progress in the understanding of JAMs, including their characteristic structural features, their molecular interactions, their cellular functions, and their contribution to a multitude of processes during vertebrate development and homeostasis.
Collapse
Affiliation(s)
- Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity", Institute of Medical Biochemistry, ZMBE, Cells-In-Motion Cluster of Excellence (EXC1003-CiM), and Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| |
Collapse
|
32
|
Terral G, Champion T, Debaene F, Colas O, Bourguet M, Wagner-Rousset E, Corvaia N, Beck A, Cianferani S. Epitope characterization of anti-JAM-A antibodies using orthogonal mass spectrometry and surface plasmon resonance approaches. MAbs 2017; 9:1317-1326. [PMID: 28933642 DOI: 10.1080/19420862.2017.1380762] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Junctional adhesion molecule-A (JAM-A) is an adherens and tight junction protein expressed by endothelial and epithelial cells and associated with cancer progression. We present here the extensive characterization of immune complexes involving JAM-A antigen and three monoclonal antibodies (mAbs), including hz6F4-2, a humanized version of anti-tumoral 6F4 mAb identified by a functional and proteomic approach in our laboratory. A specific workflow that combines orthogonal approaches has been designed to determine binding stoichiometries along with JAM-A epitope mapping determination at high resolution for these three mAbs. Native mass spectrometry experiments revealed different binding stoichiometries and affinities, with two molecules of JAM-A being able to bind to hz6F4-2 and F11 Fab, while only one JAM-A was bound to J10.4. Surface plasmon resonance indirect competitive binding assays suggested epitopes located in close proximity for hz6F4-2 and F11. Finally, hydrogen-deuterium exchange mass spectrometry was used to precisely identify epitopes for all mAbs. The results obtained by orthogonal biophysical approaches showed a clear correlation between the determined epitopes and JAM-A binding characteristics, allowing the basis for molecular recognition of JAM-A by hz6F4-2 to be definitively established for the first time. Taken together, our results highlight the power of MS-based structural approaches for epitope mapping and mAb conformational characterization.
Collapse
Affiliation(s)
- Guillaume Terral
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178 , Strasbourg , France
| | - Thierry Champion
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | - François Debaene
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178 , Strasbourg , France
| | - Olivier Colas
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | - Maxime Bourguet
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178 , Strasbourg , France
| | - Elsa Wagner-Rousset
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | - Nathalie Corvaia
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | - Alain Beck
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | - Sarah Cianferani
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178 , Strasbourg , France
| |
Collapse
|
33
|
Identification of Human Junctional Adhesion Molecule 1 as a Functional Receptor for the Hom-1 Calicivirus on Human Cells. mBio 2017; 8:mBio.00031-17. [PMID: 28196955 PMCID: PMC5312078 DOI: 10.1128/mbio.00031-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Hom-1 vesivirus was reported in 1998 following the inadvertent transmission of the animal calicivirus San Miguel sea lion virus to a human host in a laboratory. We characterized the Hom-1 strain and investigated the mechanism by which human cells could be infected. An expression library of 3,559 human plasma membrane proteins was screened for reactivity with Hom-1 virus-like particles, and a single interacting protein, human junctional adhesion molecule 1 (hJAM1), was identified. Transient expression of hJAM1 conferred susceptibility to Hom-1 infection on nonpermissive Chinese hamster ovary (CHO) cells. Virus infection was markedly inhibited when CHO cells stably expressing hJAM were pretreated with anti-hJAM1 monoclonal antibodies. Cell lines of human origin were tested for growth of Hom-1, and efficient replication was observed in HepG2, HuH7, and SK-CO15 cells. The three cell lines (of hepatic or intestinal origin) were confirmed to express hJAM1 on their surface, and clustered regularly interspaced short palindromic repeats/Cas9-mediated knockout of the hJAM1 gene in each line abolished Hom-1 propagation. Taken together, our data indicate that entry of the Hom-1 vesivirus into these permissive human cell lines is mediated by the plasma membrane protein hJAM1 as a functional receptor.IMPORTANCE Vesiviruses, such as San Miguel sea lion virus and feline calicivirus, are typically associated with infection in animal hosts. Following the accidental infection of a laboratory worker with San Miguel sea lion virus, a related virus was isolated in cell culture and named Hom-1. In this study, we found that Hom-1 could be propagated in a number of human cell lines, making it the first calicivirus to replicate efficiently in cultured human cells. Screening of a library of human cell surface membrane proteins showed that the virus could utilize human junctional adhesion molecule 1 as a receptor to enter cells and initiate replication. The Hom-1 virus presents a new system for the study of calicivirus biology and species specificity.
Collapse
|
34
|
Structural Insights into Reovirus σ1 Interactions with Two Neutralizing Antibodies. J Virol 2017; 91:JVI.01621-16. [PMID: 27928010 DOI: 10.1128/jvi.01621-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/29/2016] [Indexed: 01/09/2023] Open
Abstract
Reovirus attachment protein σ1 engages glycan receptors and junctional adhesion molecule-A (JAM-A) and is thought to undergo a conformational change during the proteolytic disassembly of virions to infectious subvirion particles (ISVPs) that accompanies cell entry. The σ1 protein is also the primary target of neutralizing antibodies. Here, we present a structural and functional characterization of two neutralizing antibodies that target σ1 of serotype 1 (T1) and serotype 3 (T3) reoviruses. The crystal structures revealed that each antibody engages its cognate σ1 protein within the head domain via epitopes distinct from the JAM-A-binding site. Surface plasmon resonance and cell-binding assays indicated that both antibodies likely interfere with JAM-A engagement by steric hindrance. To define the interplay between the carbohydrate receptor and antibody binding, we conducted hemagglutination inhibition assays using virions and ISVPs. The glycan-binding site of T1 σ1 is located in the head domain and is partly occluded by the bound Fab in the crystal structure. The T1-specific antibody inhibited hemagglutination by virions and ISVPs, probably via direct interference with glycan engagement. In contrast to T1 σ1, the carbohydrate-binding site of T3 σ1 is located in the tail domain, distal to the antibody epitope. The T3-specific antibody inhibited hemagglutination by T3 virions but not ISVPs, indicating that the antibody- and glycan-binding sites in σ1 are in closer spatial proximity on virions than on ISVPs. Our results provide direct evidence for a structural rearrangement of σ1 during virion-to-ISVP conversion and contribute new information about the mechanisms of antibody-mediated neutralization of reovirus. IMPORTANCE Virus attachment proteins mediate binding to host cell receptors, serve critical functions in cell and tissue tropism, and are often targeted by the neutralizing antibody response. The structural investigation of antibody-antigen complexes can provide valuable information for understanding the molecular basis of virus neutralization. Studies with enveloped viruses, such as HIV and influenza virus, have helped to define sites of vulnerability and guide vaccination strategies. By comparison, less is known about antibody binding to nonenveloped viruses. Here, we structurally investigated two neutralizing antibodies that bind the attachment protein σ1 of reovirus. Furthermore, we characterized the neutralization efficiency, the binding affinity for σ1, and the effect of the antibodies on reovirus receptor engagement. Our analysis defines reovirus interactions with two neutralizing antibodies, allows us to propose a mechanism by which they block virus infection, and provides evidence for a conformational change in the σ1 protein during viral cell entry.
Collapse
|
35
|
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: 1.7] [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
|
36
|
Luissint AC, Parkos CA, Nusrat A. Inflammation and the Intestinal Barrier: Leukocyte-Epithelial Cell Interactions, Cell Junction Remodeling, and Mucosal Repair. Gastroenterology 2016; 151:616-32. [PMID: 27436072 PMCID: PMC5317033 DOI: 10.1053/j.gastro.2016.07.008] [Citation(s) in RCA: 390] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/13/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
The intestinal tract is lined by a single layer of columnar epithelial cells that forms a dynamic, permeable barrier allowing for selective absorption of nutrients, while restricting access to pathogens and food-borne antigens. Precise regulation of epithelial barrier function is therefore required for maintaining mucosal homeostasis and depends, in part, on barrier-forming elements within the epithelium and a balance between pro- and anti-inflammatory factors in the mucosa. Pathologic states, such as inflammatory bowel disease, are associated with a leaky epithelial barrier, resulting in excessive exposure to microbial antigens, recruitment of leukocytes, release of soluble mediators, and ultimately mucosal damage. An inflammatory microenvironment affects epithelial barrier properties and mucosal homeostasis by altering the structure and function of epithelial intercellular junctions through direct and indirect mechanisms. We review our current understanding of complex interactions between the intestinal epithelium and immune cells, with a focus on pathologic mucosal inflammation and mechanisms of epithelial repair. We discuss leukocyte-epithelial interactions, as well as inflammatory mediators that affect the epithelial barrier and mucosal repair. Increased knowledge of communication networks between the epithelium and immune system will lead to tissue-specific strategies for treating pathologic intestinal inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
37
|
ZHAO HUISHAN, YU HEFEN, MARTIN TRACEYA, TENG XU, JIANG WENG. The role of JAM-B in cancer and cancer metastasis (Review). Oncol Rep 2016; 36:3-9. [PMID: 27121546 PMCID: PMC4899009 DOI: 10.3892/or.2016.4773] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/08/2015] [Indexed: 12/31/2022] Open
Abstract
The junctional adhesion molecule B (JAM-B) is a multifunctional transmembrane protein, which belongs to the immunoglobulin superfamily (IgSF). JAM-B is localized to cell-cell contacts and enriched at cell junctions in epithelial and endothelial cells, as well as on the surface of erythrocytes, leukocytes, and platelets. Recent research in this field has shown that JAM-B plays an important role in numerous cellular processes, such as tight junction assembly, spermatogenesis, regulation of paracellular permeability, leukocytic transmigration, angiogenesis, tumor metastasis and cell proliferation. This study provides a new research direction for the diagnosis and treatment of relevant diseases. In this review, we briefly focus on what is currently known about the structure, function, and mechanism of JAM-B, with particular emphasis on cancer.
Collapse
Affiliation(s)
- HUISHAN ZHAO
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical university, Beijing 100069, P.R. China
- Cancer Institute of Capital Medical university, Beijing 100069, P.R. China
- Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Beijing 100069, P.R. China
- Cardiff-China Medical Research Collaborative, Cardiff university School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - HEFEN YU
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical university, Beijing 100069, P.R. China
- Cancer Institute of Capital Medical university, Beijing 100069, P.R. China
- Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Beijing 100069, P.R. China
| | - TRACEY A. MARTIN
- Cardiff-China Medical Research Collaborative, Cardiff university School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - XU TENG
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical university, Beijing 100069, P.R. China
- Cancer Institute of Capital Medical university, Beijing 100069, P.R. China
- Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Beijing 100069, P.R. China
- Cardiff-China Medical Research Collaborative, Cardiff university School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - WEN G. JIANG
- Cancer Institute of Capital Medical university, Beijing 100069, P.R. China
- Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Beijing 100069, P.R. China
- Cardiff-China Medical Research Collaborative, Cardiff university School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
38
|
Babinska A, Clement CC, Swiatkowska M, Szymanski J, Shon A, Ehrlich YH, Kornecki E, Salifu MO. Development of new antiatherosclerotic and antithrombotic drugs utilizing F11 receptor (F11R/JAM-A) peptides. Biopolymers 2016; 102:322-34. [PMID: 24801754 DOI: 10.1002/bip.22503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/01/2014] [Accepted: 05/02/2014] [Indexed: 12/29/2022]
Abstract
Peptides with enhanced resistance to proteolysis, based on the amino acid sequence of the F11 receptor molecule (F11R, aka JAM-A/Junctional adhesion molecule-A), were designed, prepared, and examined as potential candidates for the development of anti-atherosclerotic and anti-thrombotic therapeutic drugs. A sequence at the N-terminal of F11R together with another sequence located in the first Ig-loop of this protein, were identified to form a steric active-site operating in the F11R-dependent adhesion between cells that express F11R molecules on their external surface. In silico modeling of the complex between two polypeptide chains with the sequences positioned in the active-site was used to generate peptide-candidates designed to inhibit homophilic interactions between surface-located F11R molecules. The two lead F11R peptides were modified with D-Arg and D-Lys at selective sites, for attaining higher stability to proteolysis in vivo. Using molecular docking experiments we tested different conformational states and the putative binding affinity between two selected D-Arg and D-Lys-modified F11R peptides and the proposed binding pocket. The inhibitory effects of the F11R peptide 2HN-(dK)-SVT-(dR)-EDTGTYTC-CONH2 on antibody-induced platelet aggregation and on the adhesion of platelets to cytokine-inflammed endothelial cells are reported in detail, and the results point out the significant potential utilization of F11R peptides for the prevention and treatment of atherosclerotic plaques and associated thrombotic events.
Collapse
Affiliation(s)
- A Babinska
- Division of Nephrology, Department of Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203; Department of Cell Biology and Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, 11203
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Mateo M, Generous A, Sinn PL, Cattaneo R. Connections matter--how viruses use cell–cell adhesion components. J Cell Sci 2016; 128:431-9. [PMID: 26046138 DOI: 10.1242/jcs.159400] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epithelium is a highly organized type of animal tissue. Except for blood and lymph vessels, epithelial cells cover the body, line its cavities in single or stratified layers and support exchange between compartments. In addition, epithelia offer to the body a barrier to pathogen invasion. To transit through or to replicate in epithelia, viruses have to face several obstacles, starting from cilia and glycocalyx where they can be neutralized by secreted immunoglobulins. Tight junctions and adherens junctions also prevent viruses to cross the epithelial barrier. However, viruses have developed multiple strategies to blaze their path through the epithelium by utilizing components of cell–cell adhesion structures as receptors. In this Commentary, we discuss how viruses take advantage of the apical junction complex to spread. Whereas some viruses quickly disrupt epithelium integrity, others carefully preserve it and use cell adhesion proteins and their cytoskeletal connections to rapidly spread laterally. This is exemplified by the hidden transmission of enveloped viruses that use nectins as receptors. Finally, several viruses that replicate preferentially in cancer cells are currently used as experimental cancer therapeutics. Remarkably, these viruses use cell adhesion molecules as receptors, probably because--to reach tumors and metastases--ncolytic viruses must efficiently traverse or break epithelia.
Collapse
|
40
|
Laminin receptor is an interacting partner for viral outer capsid protein VP5 in grass carp reovirus infection. Virology 2016; 490:59-68. [PMID: 26848829 DOI: 10.1016/j.virol.2016.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 11/19/2015] [Accepted: 01/21/2016] [Indexed: 11/23/2022]
Abstract
Grass carp reovirus (GCRV) is responsible for viral hemorrhagic disease in cultured grass carp Ctenopharyngon idellus. Through yeast two-hybrid screen, laminin receptor (LamR) was identified as a potential interacting partner for the outer capsid protein VP5 of GCRV. We cloned and sequenced the gene encoding grass carp LamR. Viral attachment assay demonstrated the involvement of membrane-associated LamR in GCRV infection. Solid-phase overlay assays demonstrated that GCRV interacted with GST-tagged LamR in vitro. In contrast to VP7, GST-tagged VP5 was shown to associate with LamR in both pull-down and solid-phase blot overlay assays. With the reduction of LamR expression in CIK cells achieved by RNAi, remarkably reduced infection efficiency of GCRV was observed. CIK cells pretreated with polyclonal antibody against LamR resulted in dose-dependent inhibition of GCRV infection. These results collectively indicated that grass carp LamR was involved in GCRV infection by interacting with viral outer capsid protein VP5.
Collapse
|
41
|
Liu C, Wang M, Jiang S, Wang L, Chen H, Liu Z, Qiu L, Song L. A novel junctional adhesion molecule A (CgJAM-A-L) from oyster (Crassostrea gigas) functions as pattern recognition receptor and opsonin. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 55:211-220. [PMID: 26434620 DOI: 10.1016/j.dci.2015.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 06/05/2023]
Abstract
Junctional adhesion molecule (JAM), a subfamily of immunoglobulin superfamily (IgSF) with a couple of immunoglobulin domains, can act as regulator in homeostasis and inflammation of vertebrates. In the present study, a structural homolog of JAM-A (designated CgJAM-A-L) was screened out from oyster, Crassostrea gigas, through a search of JAM-A D1 domain (N-terminal Ig domain in JAM-A). The cDNA of CgJAM-A-L was of 1188 bp encoding a predicted polypeptide of 395 amino acids. The immunoreactive area of CgJAM-A-L mainly distributed over the plasma membrane of hemocytes. After Vibro splendidus or tumor necrosis factor (CgTNF-1) stimulation, the mRNA transcripts of CgJAM-A-L in hemocytes increased significantly by 4.46-fold and 9.00-fold (p < 0.01) of those in control group, respectively. The recombinant CgJAM-A-L protein (rCgJAM-A-L) could bind multiple PAMPs including lipopolysaccharides (LPS), peptidoglycan (PGN), lipoteichoic acid (LTA), mannose (MAN), β-glucan (GLU) and poly(I:C), and various microorganisms including Micrococcus luteus, Staphylococcus aureus, Escherichia coli, Vibro anguillarum, V. splendidus, Pastoris pastoris and Yarrowia lipolytica. The phagocytic rates of oyster hemocytes towards Gram-negative bacteria V. anguillarum and yeast P. pastoris were significantly enhanced after the incubation of rCgJAM-A-L, and even increased more significantly after the pre-incubation of rCgJAM-A-L with microbes (p < 0.01). The results collectively indicated that CgJAM-A-L functioned as an important pattern recognition receptor (PRR) and opsonin in the immune defense against invading pathogen in oyster. Moreover, as the most primitive specie with homolog of JAMs, the information of CgJAM-A-L in oyster would provide useful clues for the evolutionary study of JAMs and immunoglobulins.
Collapse
Affiliation(s)
- Conghui Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengqiang Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Shuai Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Lingling Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Hao Chen
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaoqun Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Linsheng Song
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
42
|
Bhella D. The role of cellular adhesion molecules in virus attachment and entry. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140035. [PMID: 25533093 PMCID: PMC4275905 DOI: 10.1098/rstb.2014.0035] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
As obligate intracellular parasites, viruses must traverse the host-cell plasma membrane to initiate infection. This presents a formidable barrier, which they have evolved diverse strategies to overcome. Common to all entry pathways, however, is a mechanism of specific attachment to cell-surface macromolecules or ‘receptors’. Receptor usage frequently defines viral tropism, and consequently, the evolutionary changes in receptor specificity can lead to emergence of new strains exhibiting altered pathogenicity or host range. Several classes of molecules are exploited as receptors by diverse groups of viruses, including, for example, sialic acid moieties and integrins. In particular, many cell-adhesion molecules that belong to the immunoglobulin-like superfamily of proteins (IgSF CAMs) have been identified as viral receptors. Structural analysis of the interactions between viruses and IgSF CAM receptors has not shown binding to specific features, implying that the Ig-like fold may not be key. Both proteinaceous and enveloped viruses exploit these proteins, however, suggesting convergent evolution of this trait. Their use is surprising given the usually occluded position of CAMs on the cell surface, such as at tight junctions. Nonetheless, the reason for their widespread involvement in virus entry most probably originates in their functional rather than structural characteristics.
Collapse
Affiliation(s)
- David Bhella
- Medical Research Council-University of Glasgow Centre for Virus Research, Sir Michael Stoker Building, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK
| |
Collapse
|
43
|
Mohamed A, Johnston RN, Shmulevitz M. Potential for Improving Potency and Specificity of Reovirus Oncolysis with Next-Generation Reovirus Variants. Viruses 2015; 7:6251-78. [PMID: 26633466 PMCID: PMC4690860 DOI: 10.3390/v7122936] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 11/10/2015] [Accepted: 11/18/2015] [Indexed: 12/16/2022] Open
Abstract
Viruses that specifically replicate in tumor over normal cells offer promising cancer therapies. Oncolytic viruses (OV) not only kill the tumor cells directly; they also promote anti-tumor immunotherapeutic responses. Other major advantages of OVs are that they dose-escalate in tumors and can be genetically engineered to enhance potency and specificity. Unmodified wild type reovirus is a propitious OV currently in phase I–III clinical trials. This review summarizes modifications to reovirus that may improve potency and/or specificity during oncolysis. Classical genetics approaches have revealed reovirus variants with improved adaptation towards tumors or with enhanced ability to establish specific steps of virus replication and cell killing among transformed cells. The recent emergence of a reverse genetics system for reovirus has provided novel strategies to fine-tune reovirus proteins or introduce exogenous genes that could promote oncolytic activity. Over the next decade, these findings are likely to generate better-optimized second-generation reovirus vectors and improve the efficacy of oncolytic reotherapy.
Collapse
Affiliation(s)
- Adil Mohamed
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Randal N Johnston
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Maya Shmulevitz
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
44
|
Assembly and function of claudins: Structure–function relationships based on homology models and crystal structures. Semin Cell Dev Biol 2015; 42:3-12. [DOI: 10.1016/j.semcdb.2015.04.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 01/12/2023]
|
45
|
Zhang T, Liu T, Shao J, Sheng C, Hong Y, Ying W, Xia W. Antioxidant protects blood-testis barrier against synchrotron radiation X-ray-induced disruption. SPERMATOGENESIS 2015; 5:e1009313. [PMID: 26413412 DOI: 10.1080/21565562.2015.1009313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/07/2015] [Accepted: 01/15/2015] [Indexed: 01/06/2023]
Abstract
Synchrotron radiation (SR) X-ray has wide biomedical applications including high resolution imaging and brain tumor therapy due to its special properties of high coherence, monochromaticity and high intensity. However, its interaction with biological tissues remains poorly understood. In this study, we used the rat testis as a model to investigate how SR X-ray would induce tissue responses, especially the blood-testis barrier (BTB) because BTB dynamics are critical for spermatogenesis. We irradiated the male gonad with increasing doses of SR X-ray and obtained the testicles 1, 10 and 20 d after the exposures. The testicle weight and seminiferous tubule diameter reduced in a dose- and time-dependent manner. Cryosections of testes were stained with tight junction (TJ) component proteins such as occludin, claudin-11, JAM-A and ZO-1. Morphologically, increasing doses of SR X-ray consistently induced developing germ cell sloughing from the seminiferous tubules, accompanied by shrinkage of the tubules. Interestingly, TJ constituent proteins appeared to be induced by the increasing doses of SR X-ray. Up to 20 d after SR X-ray irradiation, there also appeared to be time-dependent changes on the steady-state level of these protein exhibiting differential patterns at 20-day after exposure, with JAM-A/claudin-11 still being up-regulated whereas occludin/ZO-1 being down-regulated. More importantly, the BTB damage induced by 40 Gy of SR X-ray could be significantly attenuated by antioxidant N-Acetyl-L-Cysteine (NAC) at a dose of 125 mg/kg. Taken together, our studies characterized the changes of TJ component proteins after SR X-ray irradiation, illustrating the possible protective effects of antioxidant NAC to BTB integrity.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Laboratory of Oncogenes and Related Genes; Renji-Med X Clinical Stem Cell Research Center; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University ; Shanghai, China ; School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University ; Shanghai, China
| | - Tengyuan Liu
- State Key Laboratory of Oncogenes and Related Genes; Renji-Med X Clinical Stem Cell Research Center; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University ; Shanghai, China ; School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University ; Shanghai, China
| | - Jiaxiang Shao
- State Key Laboratory of Oncogenes and Related Genes; Renji-Med X Clinical Stem Cell Research Center; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University ; Shanghai, China ; School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University ; Shanghai, China
| | - Caibin Sheng
- School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University ; Shanghai, China
| | - Yunyi Hong
- School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University ; Shanghai, China
| | - Weihai Ying
- School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University ; Shanghai, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes; Renji-Med X Clinical Stem Cell Research Center; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University ; Shanghai, China ; School of Biomedical Engineering & Med-X Research Institute; Shanghai Jiao Tong University ; Shanghai, China
| |
Collapse
|
46
|
Structure of Serotype 1 Reovirus Attachment Protein σ1 in Complex with Junctional Adhesion Molecule A Reveals a Conserved Serotype-Independent Binding Epitope. J Virol 2015; 89:6136-40. [PMID: 25810543 DOI: 10.1128/jvi.00433-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/17/2015] [Indexed: 01/02/2023] Open
Abstract
Mammalian orthoreoviruses use glycans and junctional adhesion molecule A (JAM-A) as attachment receptors. We determined the structure of serotype 1 reovirus attachment protein σ1 alone and in complex with JAM-A. Comparison with the structure of serotype 3 reovirus σ1 bound to JAM-A reveals that both σ1 proteins engage JAM-A with similar affinities and via conserved binding epitopes. Thus, σ1-JAM-A interactions are unlikely to explain the differences in pathogenesis displayed by these reovirus serotypes.
Collapse
|
47
|
RNA interference mediated JAM-A gene silencing promotes human epidermal stem cell proliferation. Hum Cell 2014; 28:73-80. [DOI: 10.1007/s13577-013-0087-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 12/23/2013] [Indexed: 10/24/2022]
|
48
|
Monteiro AC, Luissint AC, Sumagin R, Lai C, Vielmuth F, Wolf MF, Laur O, Reiss K, Spindler V, Stehle T, Dermody TS, Nusrat A, Parkos CA. Trans-dimerization of JAM-A regulates Rap2 and is mediated by a domain that is distinct from the cis-dimerization interface. Mol Biol Cell 2014; 25:1574-85. [PMID: 24672055 PMCID: PMC4019489 DOI: 10.1091/mbc.e14-01-0018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Junctional adhesion molecule-A (JAM-A) is a tight junction–associated signaling protein that homodimerizes across cells at a unique motif to activate the small GTPase Rap2, previously implicated in the regulation of barrier function. JAM-A may therefore act as a barrier-inducing molecular switch that is activated when cells become confluent. Junctional adhesion molecule-A (JAM-A) is a tight junction–associated signaling protein that regulates epithelial cell proliferation, migration, and barrier function. JAM-A dimerization on a common cell surface (in cis) has been shown to regulate cell migration, and evidence suggests that JAM-A may form homodimers between cells (in trans). Indeed, transfection experiments revealed accumulation of JAM-A at sites between transfected cells, which was lost in cells expressing cis- or predicted trans-dimerization null mutants. Of importance, microspheres coated with JAM-A containing alanine substitutions to residues 43NNP45 (NNP-JAM-A) within the predicted trans-dimerization site did not aggregate. In contrast, beads coated with cis-null JAM-A demonstrated enhanced clustering similar to that observed with wild-type (WT) JAM-A. In addition, atomic force microscopy revealed decreased association forces in NNP-JAM-A compared with WT and cis-null JAM-A. Assessment of effects of JAM-A dimerization on cell signaling revealed that expression of trans- but not cis-null JAM-A mutants decreased Rap2 activity. Furthermore, confluent cells, which enable trans-dimerization, had enhanced Rap2 activity. Taken together, these results suggest that trans-dimerization of JAM-A occurs at a unique site and with different affinity compared with dimerization in cis. Trans-dimerization of JAM-A may thus act as a barrier-inducing molecular switch that is activated when cells become confluent.
Collapse
Affiliation(s)
- Ana C Monteiro
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Anny-Claude Luissint
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Ronen Sumagin
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Caroline Lai
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Franziska Vielmuth
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Mattie F Wolf
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Oskar Laur
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Kerstin Reiss
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076 Tübingen, Germany
| | - Volker Spindler
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Thilo Stehle
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, TN 37232Interfaculty Institute of Biochemistry, University of Tübingen, D-72076 Tübingen, Germany
| | - Terence S Dermody
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, TN 37232Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Charles A Parkos
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
49
|
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: 7.2] [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
|
50
|
Garrido-Urbani S, Bradfield PF, Imhof BA. Tight junction dynamics: the role of junctional adhesion molecules (JAMs). Cell Tissue Res 2014; 355:701-15. [DOI: 10.1007/s00441-014-1820-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/16/2014] [Indexed: 12/27/2022]
|