51
|
Yang H, Xiong H, Mi K, Zhang Y, Zhang X, Chen G. The surface syndecan protein from Macrobrachium rosenbergii could function as mediator in bacterial infections. FISH & SHELLFISH IMMUNOLOGY 2020; 96:62-68. [PMID: 31704203 DOI: 10.1016/j.fsi.2019.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/26/2019] [Accepted: 11/04/2019] [Indexed: 06/10/2023]
Abstract
Due to the aquatic animal pathogens are numerous and specific, the pathogen invasion mechanisms are more complicated. The cell surface receptors play vital roles to understand these mechanisms. Syndecan is a cell surface protein and could function as a receptor involved bacteria and virus infections. But there are few studies on the function of syndecan in shrimp and their interaction with aquatic bacterial pathogens. In the present study, we identified a syndecan receptor gene from Macrobrachium rosenbergii and analyzed its functions during the bacterial infections. The MrSDC was expressed in various tissues and presented a constitutive expression distribution except in eyestalk. Recombinant MrSDC-his tag protein was expressed in the E. coli BL21 with pET30a/MrSDC plasmid and exhibited a broad bacterial binding activities. The inhibition of MrSDC expression by dsRNA interference and antibody blocked could significantly reduce the number of Aeromonas hydrophila in hepatopancreas compared with the control. The overexpression of MrSDC by mRNA injection could significantly increase the number of A. hydrophila. In addition, the functional role of syndecan heparan sulfate chains in bacterial recognition was also studied. After extra injection of heparan sulfate in vivo, the bacterial numbers and accumulative mortality of M. rosenbergii were significantly higher than control groups and exhibit a dose effect. All these data could indicate that the cell surface syndecan protein could function as mediator in bacterial infections by the heparan sulfate chains. Our present study will provide new insights into the functions of shrimp syndecan.
Collapse
Affiliation(s)
- Hui Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Haoran Xiong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Kaihang Mi
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yingying Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Xiaojun Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Guohong Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
52
|
He L, Xu H, Ye F, Yu H, Lu Y, Yin H, Zhao X, Zhu Q, Wang Y. Expression Pattern of Sulf1 and Sulf2 in Chicken Tissues and Characterization of Their Expression During Different Periods in Skeletal Muscle Satellite Cells. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2020. [DOI: 10.1590/1806-9061-2019-1165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- L He
- Sichuan Agricultural University, China
| | - H Xu
- Sichuan Agricultural University, China
| | - F Ye
- Sichuan Agricultural University, China
| | - H Yu
- Sichuan Agricultural University, China
| | - Y Lu
- Sichuan Agricultural University, China
| | - H Yin
- Sichuan Agricultural University, China
| | - X Zhao
- Sichuan Agricultural University, China
| | - Q Zhu
- Sichuan Agricultural University, China
| | - Y Wang
- Sichuan Agricultural University, China
| |
Collapse
|
53
|
Hudák A, Kusz E, Domonkos I, Jósvay K, Kodamullil AT, Szilák L, Hofmann-Apitius M, Letoha T. Contribution of syndecans to cellular uptake and fibrillation of α-synuclein and tau. Sci Rep 2019; 9:16543. [PMID: 31719623 PMCID: PMC6851098 DOI: 10.1038/s41598-019-53038-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 10/28/2019] [Indexed: 11/09/2022] Open
Abstract
Scientific evidence suggests that α-synuclein and tau have prion-like properties and that prion-like spreading and seeding of misfolded protein aggregates constitutes a central mechanism for neurodegeneration. Heparan sulfate proteoglycans (HSPGs) in the plasma membrane support this process by attaching misfolded protein fibrils. Despite of intense studies, contribution of specific HSPGs to seeding and spreading of α-synuclein and tau has not been explored yet. Here we report that members of the syndecan family of HSPGs mediate cellular uptake of α-synuclein and tau fibrils via a lipid-raft dependent and clathrin-independent endocytic route. Among syndecans, the neuron predominant syndecan-3 exhibits the highest affinity for both α-synuclein and tau. Syndecan-mediated internalization of α-synuclein and tau depends heavily on conformation as uptake via syndecans start to dominate once fibrils are formed. Overexpression of syndecans, on the other hand, reduces cellular uptake of monomeric α-synuclein and tau, yet exerts a fibril forming effect on both proteins. Data obtained from syndecan overexpressing cellular models presents syndecans, especially the neuron predominant syndecan-3, as important mediators of seeding and spreading of α-synuclein and tau and reveal how syndecans contribute to fundamental molecular events of α-synuclein and tau pathology.
Collapse
Affiliation(s)
| | | | - Ildikó Domonkos
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Katalin Jósvay
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Alpha Tom Kodamullil
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53754, Germany
| | - László Szilák
- Szilak Laboratories, Bioinformatics and Molecule-Design, Szeged, H-6723, Hungary
| | - Martin Hofmann-Apitius
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53754, Germany
| | | |
Collapse
|
54
|
Mechanism of anchorage-independency and tumor formation of cancer cells: possible involvement of cell membrane-bound laminin-332. Cell Tissue Res 2019; 379:255-259. [PMID: 31705213 DOI: 10.1007/s00441-019-03114-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/22/2019] [Indexed: 02/08/2023]
Abstract
Cancer cells are characterized by anchorage-independency and tumor formation. Involvement of laminin-332 in the pathogenesis of cancer has also been reported. I present a theory that can explain these characteristics together. Proliferating keratinocytes in wound healing produce and deposit laminin-332, which is shown in the provisional basement membrane of a wound. In association with wound closure, expression of LG4/5 domain on the α3 chain of laminin-332 disappears, implicating cleavage of LG4/5 domain. LG4/5 domain expression indicates that laminin-332 prior to the cleavage is bound to the cell membrane, because LG4/5 domain is a cell binding site. In this binding, heparan sulfate proteoglycan on the cell surface seems to be the acceptor for LG4/5 domain. I named this laminin "cell membrane-bound laminin-332" (ML332). ML332 would then bind to integrin α3β1 via LG1-3 domain, the integrin binding site, and activate FAK and the following Ras/MAPK pathway. Therefore, ML332 eliminates the need for proliferating keratinocytes to bind to processed laminin-332 secreted and deposited into the basement membrane for their proliferation (anchorage-independency). This may hold true of every proliferating epithelial cell, either benign or malignant. Whereas wound closure deprives keratinocytes of anchorage-independency, such events do not occur in cancer cells, and cancer cells maintain anchorage-independency. In the basement membrane formation by epithelial cells, short arms of laminin-332 anchored to the cell membrane bind each other and generate a meshwork polymer. This is the three-arm interaction model. In a similar manner, short-arm interactions between adjacent cancer cells may occur and induce tumor formation.
Collapse
|
55
|
Duplancic R, Roguljic M, Puhar I, Vecek N, Dragun R, Vukojevic K, Saraga-Babic M, Kero D. Syndecans and Enzymes for Heparan Sulfate Biosynthesis and Modification Differentially Correlate With Presence of Inflammatory Infiltrate in Periodontitis. Front Physiol 2019; 10:1248. [PMID: 31611818 PMCID: PMC6773826 DOI: 10.3389/fphys.2019.01248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/12/2019] [Indexed: 01/20/2023] Open
Abstract
Periodontitis is a common degenerative disease initiated by the bacteria in subgingival biofilm. The exposure to bacterial biofilm triggers host inflammatory response whose dysregulation is ultimately responsible for the destruction of hard and soft periodontal tissues resulting in tooth loss. To date, significant effort has been invested in the research of the involvement of host cells and inflammatory mediators in regulation of inflammatory response in periodontitis. Syndecans (Sdcs) belong to a four-member family of heparan sulfate proteoglycans (HSPGs). Sdcs are compound molecules comprised of the core protein to which several heparan sulfate (HS) glycosaminoglycan (GAG) chains are attached. The role of Sdcs in pathogenesis of periodontitis is poorly investigated despite the numerous reports from experimental studies about the critical involvement of these factors in modulation of various aspects of inflammatory response, such as the formation of inflammatory mediators gradients, leukocyte recruitment and extracellular matrix remodeling in resolution of inflammation. Most of these functions of Sdcs are HS-related and, thus, dependent upon the structure of HS. This, in turn, is determined by the combinatorial action of enzymes for biosynthesis and modification of HS such as exostosis (EXTs), sulfotransferases (NDSTs), and heparanase 1 (HPSE1). The data presented in this study clearly indicate that some Sdcs display different expression profiles in healthy and diseased periodontal tissue. Additionally, the differences in expression profiles of HS GAG biosynthesis and modification enzymes (EXTs, NDSTs, and HPSE1) in healthy and diseased periodontal tissue imply that changes in HS GAG content and structure might also take place during periodontitis. Most notably, expression profiles of Sdcs, EXTs, NDSTs, and HPSE1 differentially correlate with the presence of inflammatory infiltrate in healthy and diseased periodontal tissue, which might imply that these factors could also be involved in modulation of inflammatory response in periodontitis.
Collapse
Affiliation(s)
- Roko Duplancic
- Study Programme of Dental Medicine, School of Medicine, University of Split, Split, Croatia
| | - Marija Roguljic
- Department of Oral Pathology and Periodontology, Study Programme of Dental Medicine, School of Medicine, University of Split, Split, Croatia
| | - Ivan Puhar
- Department of Periodontology, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Nika Vecek
- Study Programme of Dental Medicine, School of Medicine, University of Split, Split, Croatia
| | - Ruzica Dragun
- Study Programme of Dental Medicine, School of Medicine, University of Split, Split, Croatia
| | - Katarina Vukojevic
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Split, Croatia.,Laboratory for Early Human Development, School of Medicine, University of Split, Split, Croatia
| | - Mirna Saraga-Babic
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Split, Croatia.,Laboratory for Early Human Development, School of Medicine, University of Split, Split, Croatia
| | - Darko Kero
- Study Programme of Dental Medicine, School of Medicine, University of Split, Split, Croatia.,Laboratory for Early Human Development, School of Medicine, University of Split, Split, Croatia
| |
Collapse
|
56
|
Li D, Li X, Zhou WL, Huang Y, Liang X, Jiang L, Yang X, Sun J, Li Z, Han WD, Wang W. Genetically engineered T cells for cancer immunotherapy. Signal Transduct Target Ther 2019; 4:35. [PMID: 31637014 PMCID: PMC6799837 DOI: 10.1038/s41392-019-0070-9] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
T cells in the immune system protect the human body from infection by pathogens and clear mutant cells through specific recognition by T cell receptors (TCRs). Cancer immunotherapy, by relying on this basic recognition method, boosts the antitumor efficacy of T cells by unleashing the inhibition of immune checkpoints and expands adaptive immunity by facilitating the adoptive transfer of genetically engineered T cells. T cells genetically equipped with chimeric antigen receptors (CARs) or TCRs have shown remarkable effectiveness in treating some hematological malignancies, although the efficacy of engineered T cells in treating solid tumors is far from satisfactory. In this review, we summarize the development of genetically engineered T cells, outline the most recent studies investigating genetically engineered T cells for cancer immunotherapy, and discuss strategies for improving the performance of these T cells in fighting cancers.
Collapse
Affiliation(s)
- Dan Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xue Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Wei-Lin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Yong Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xiao Liang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
- Department of Medical Oncology, Cancer Center, West China Hospital, West China Medical School, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Lin Jiang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xiao Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Jie Sun
- Department of Cell Biology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058 Zhejiang, China
- Institute of Hematology, Zhejiang University & Laboratory of Stem cell and Immunotherapy Engineering, 310058 Zhejing, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, 200032 Shanghai, China
- CARsgen Therapeutics, 200032 Shanghai, China
| | - Wei-Dong Han
- Molecular & Immunological Department, Biotherapeutic Department, Chinese PLA General Hospital, No. 28 Fuxing Road, 100853 Beijing, China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| |
Collapse
|
57
|
Shibue T, Reinhardt F, Weinberg RA. Syndecan-Mediated Ligation of ECM Proteins Triggers Proliferative Arrest of Disseminated Tumor Cells. Cancer Res 2019; 79:5944-5957. [PMID: 31481497 DOI: 10.1158/0008-5472.can-19-1165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/18/2019] [Accepted: 08/27/2019] [Indexed: 11/16/2022]
Abstract
Systemic dissemination of tumor cells often begins long before the development of overt metastases, revealing the inefficient nature of the metastatic process. Thus, already at the time of initial clinical presentation, many patients with cancer harbor a myriad disseminated tumor cells (DTC) throughout the body, most of which are found as mitotically quiescent solitary cells. This indicates that the majority of DTCs fail, for still unknown reasons, to initiate rapid proliferation after entering foreign tissue, which likely contributes significantly to the inefficiency of metastasis formation. Here, we showed that extracellular matrix (ECM) components of the host parenchyma prevented proliferation of DTCs that had recently infiltrated foreign tissue by binding to syndecan receptors expressed on the surface of these cells. This led to the recruitment of the Par-3:Par-6:atypical PKC protein complex, a critical regulator of cell polarity, to the plasma membrane and release of Par-1 kinase into the cytosol. Cytosolic Par-1 bound, phosphorylated, and inactivated KSR scaffolding proteins ultimately inhibited Ras/ERK signaling and, in turn, cell proliferation. Inhibition of the syndecan-mediated signaling restored the proliferation of otherwise dormant DTCs, enabling these cells to efficiently colonize foreign tissues. Intriguingly, naturally aggressive cancer cells overcame the antiproliferative effect of syndecan-mediated signaling either by shutting down this signaling pathway or by activating a proproliferative signaling pathway that works independent of syndecan-mediated signaling. Collectively, these observations indicate that the proliferative arrest of DTCs is attributable, in part, to the syndecan-mediated ligation of ECM proteins. SIGNIFICANCE: This study identifies a novel signaling pathway that regulates the proliferative dormancy of individual disseminated tumor cells.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/23/5944/F1.large.jpg.
Collapse
Affiliation(s)
- Tsukasa Shibue
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts.,Ludwig Center for Molecular Oncology at MIT, Cambridge, Massachusetts
| | - Ferenc Reinhardt
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts. .,Ludwig Center for Molecular Oncology at MIT, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
58
|
Zandonadi FS, Castañeda Santa Cruz E, Korvala J. New SDC function prediction based on protein-protein interaction using bioinformatics tools. Comput Biol Chem 2019; 83:107087. [PMID: 31351242 DOI: 10.1016/j.compbiolchem.2019.107087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/13/2019] [Accepted: 06/23/2019] [Indexed: 12/11/2022]
Abstract
The precise roles for SDC have been complex to specify. Assigning and reanalyzing protein and peptide identification to novel protein functions is one of the most important challenges in postgenomic era. Here, we provide SDC molecular description to support, contextualize and reanalyze the corresponding protein-protein interaction (PPI). From SDC-1 data mining, we discuss the potential of bioinformatics tools to predict new biological rules of SDC. Using these methods, we have assembled new possibilities for SDC biology from PPI data, once, the understanding of biology complexity cannot be capture from one simple question.
Collapse
Affiliation(s)
- Flávia S Zandonadi
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Departamento de Química Analítica, Universidade de Campinas, UNICAMP, Campinas, SP, Brazil.
| | - Elisa Castañeda Santa Cruz
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Departamento de Química Analítica, Universidade de Campinas, UNICAMP, Campinas, SP, Brazil
| | - Johanna Korvala
- Cancer and Translational Medicine Research Unit, Biocenter Oulu and Faculty of Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
59
|
Eustace AD, McNaughton EF, King S, Kehoe O, Kungl A, Mattey D, Nobbs AH, Williams N, Middleton J. Soluble syndecan-3 binds chemokines, reduces leukocyte migration in vitro and ameliorates disease severity in models of rheumatoid arthritis. Arthritis Res Ther 2019; 21:172. [PMID: 31300004 PMCID: PMC6625118 DOI: 10.1186/s13075-019-1939-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/10/2019] [Indexed: 01/04/2023] Open
Abstract
Background Syndecans are heparan sulfate proteoglycans that occur in membrane-bound or soluble forms. Syndecan-3, the least well-characterised of the syndecan family, is highly expressed on synovial endothelial cells in rheumatoid arthritis patients. Here, it binds pro-inflammatory chemokines with evidence for a role in chemokine presentation and leukocyte trafficking into the joint, promoting the inflammatory response. In this study, we explored the role of soluble syndecan-3 as a binder of chemokines and as an anti-inflammatory and therapeutic molecule. Methods A human monocytic cell line and CD14+ PBMCs were utilised in both Boyden chamber and trans-endothelial migration assays. Soluble syndecan-3 was tested in antigen-induced and collagen-induced in vivo arthritis models in mice. ELISA and isothermal fluorescence titration assays assessed the binding affinities. Syndecan-3 expression was identified by flow cytometry and PCR, and levels of shedding by ELISA. Results Using in vitro and in vivo models, soluble syndecan-3 inhibited leukocyte migration in vitro in response to CCL7 and its administration in murine models of rheumatoid arthritis reduced histological disease severity. Using isothermal fluorescence titration, the binding affinity of soluble syndecan-3 to inflammatory chemokines CCL2, CCL7 and CXCL8 was determined, revealing little difference, with Kds in the low nM range. TNFα increased cell surface expression and shedding of syndecan-3 from cultured human endothelial cells. Furthermore, soluble syndecan-3 occurred naturally in the sera of patients with rheumatoid arthritis and periodontitis, and its levels correlated with syndecan-1. Conclusions This study shows that the addition of soluble syndecan-3 may represent an alternative therapeutic approach in inflammatory disease. Electronic supplementary material The online version of this article (10.1186/s13075-019-1939-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrew D Eustace
- Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| | - Emily F McNaughton
- Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| | - Sophie King
- Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| | - Oksana Kehoe
- Leopold Muller Arthritis Research Centre, Medical School, RJAH Orthopaedic Hospital, ISTM, Keele University, Oswestry, UK
| | - Andreas Kungl
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Humboldtstrasse 46, A-8010, Graz, Austria
| | - Derek Mattey
- Staffordshire Rheumatology Centre, Haywood Hospital, Stoke-on-Trent, UK
| | - Angela H Nobbs
- Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK.
| | - Neil Williams
- School of Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, BS8 1TD, Bristol, UK
| | - Jim Middleton
- Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| |
Collapse
|
60
|
Reyes I, Reyes N, Suriano R, Iacob C, Suslina N, Policastro A, Moscatello A, Schantz S, Tiwari RK, Geliebter J. Gene expression profiling identifies potential molecular markers of papillary thyroid carcinoma. Cancer Biomark 2019; 24:71-83. [PMID: 30614796 DOI: 10.3233/cbm-181758] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Thyroid cancer is the most common endocrine malignancy worldwide, with the predominant form papillary thyroid carcinoma (PTC) representing approximately 80% of cases. OBJECTIVE This study was addressed to identify potential genes and pathways involved in the pathogenesis of PTC and potential novel biomarkers for this disease. METHODS Gene expression profiling was carried out by DNA microarray technology. Validation of microarray data by qRT-PCR, western blot, and enzyme linked immunosorbent assay was also performed in a selected set of genes and gene products, with the potential to be used as diagnostic or prognostic biomarkers, such as those associated with cell adhesion, extracellular matrix (ECM) remodeling and immune/inflammatory response. RESULTS In this study we found that upregulation of extracellular activities, such as proteoglycans, ECM-receptor interaction, and cell adhesion molecules, were the most prominent feature of PTC. Significantly over-expressed genes included SDC1 (syndecan 1), SDC4 (syndecan 4), KLK7 (kallikrein-related peptidase 7), KLK10 (kallikrein-related peptidase 10), SLPI (secretory leukocyte peptidase inhibitor), GDF15 (growth/differentiation factor-15), ALOX5 (arachidonate 5-lipoxygenase), SFRP2 (secreted Frizzled-related protein 2), among others. Further, elevated KLK10 levels were detected in patients with PTC. Many of these genes belong to KEGG pathway "Proteoglycans in cancer". CONCLUSIONS Using DNA microarray analysis allowed the identification of genes and pathways with known important roles in malignant transformation, and also the discovery of novel genes that may be potential biomarkers for PTC.
Collapse
Affiliation(s)
- Ismael Reyes
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Niradiz Reyes
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA.,Genetics and Molecular Biology Research Group, University of Cartagena, Cartagena, Colombia
| | | | - Codrin Iacob
- New York Eye and Ear Infirmary, New York, NY, USA
| | - Nina Suslina
- New York Eye and Ear Infirmary, New York, NY, USA
| | - Anthony Policastro
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Augustine Moscatello
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | | | - Raj K Tiwari
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Jan Geliebter
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
61
|
Bleck D, Ma L, Erdene-Bymbadoo L, Brinks R, Schneider M, Tian L, Pongratz G. Introduction and validation of a new semi-automated method to determine sympathetic fiber density in target tissues. PLoS One 2019; 14:e0217475. [PMID: 31141555 PMCID: PMC6541301 DOI: 10.1371/journal.pone.0217475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 05/12/2019] [Indexed: 12/20/2022] Open
Abstract
In recent years, the role of sympathetic nervous fibers in chronic inflammation has become increasingly evident. At the onset of inflammation, sympathetic activity is increased in the affected tissue. However, sympathetic fibers are largely absent from chronically inflamed tissue. Apparently, there is a very dynamic relationship between sympathetic innervation and the immune system in areas of inflammation, and hence a rapid and easy method for quantification of nerve fiber density of target organs is of great value to answer potential research questions. Currently, nervous fiber densities are either determined by tedious manual counting, which is not suitable for high throughput approaches, or by expensive automated processes relying on specialized software and high-end microscopy equipment. Usually, tyrosine hydroxylase (TH) is used as the marker for sympathetic fibers. In order to overcome the current quantification bottleneck with a cost-efficient alternative, an automated process was established and compared to the classic manual approach of counting TH-positive sympathetic fibers. Since TH is not exclusively expressed on sympathetic fibers, but also in a number of catecholamine-producing cells, a prerequisite for automated determination of fiber densities is to reliably distinct between cells and fibers. Therefore, an additional staining using peripherin exclusively expressed in nervous fibers as a secondary marker was established. Using this novel approach, we studied the spleens from a syndecan-3 knockout (SDC3KO) mouse line, and demonstrated equal results on SNS fiber density for both manual and automated counts (Manual counts: wildtype: 22.57 +/- 11.72 fibers per mm2; ko: 31.95 +/- 18.85 fibers per mm2; p = 0.05; Automated counts: wildtype: 31.6 +/- 18.98 fibers per mm2; ko: 45.49 +/- 19.65 fibers per mm2; p = 0.02). In conclusion, this new and simple method can be used as a high-throughput approach to reliably and quickly estimate SNS nerve fiber density in target tissues.
Collapse
Affiliation(s)
- Dennis Bleck
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Li Ma
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lkham Erdene-Bymbadoo
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ralph Brinks
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Matthias Schneider
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Li Tian
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine and Translational Medicine, Department of Physiology, Faculty of Medicine, University of Tartu, Tartu, Estonia
- * E-mail: (GP); (LT)
| | - Georg Pongratz
- Hiller Research Center Rheumatology at University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- * E-mail: (GP); (LT)
| |
Collapse
|
62
|
Prognostic role of immune infiltrates in breast ductal carcinoma in situ. Breast Cancer Res Treat 2019; 177:17-27. [PMID: 31134489 DOI: 10.1007/s10549-019-05272-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/06/2019] [Indexed: 01/13/2023]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) of the breast is often regarded as a non-obligate precursor to invasive breast carcinoma but current diagnostic tools are unable to accurately predict the invasive potential of DCIS. Infiltration of immune cells into the tumour and its microenvironment is often an early event at the site of tumourigenesis. These immune infiltrates may be potential predictive and/or prognostic biomarkers for DCIS. This review aims to discuss recent findings pertaining to the potential prognostic significance of immune infiltrates as well as their evaluation in DCIS. METHODS A literature search on PubMed was conducted up to 28th January 2019. Search terms used were "DCIS", "ductal carcinoma in situ", "immune", "immunology", "TIL", "TIL assessment", and "tumour-infiltrating lymphocyte". Search filters for "Most Recent" and "English" were applied. Information from published papers related to the research topic were synthesised and summarised for this review. RESULTS Studies have revealed that immune infiltrates play a role in the biology and microenvironment of DCIS, as well as treatment response. There is currently no consensus on the evaluation of TILs in DCIS for clinical application. CONCLUSIONS This review highlights the recent findings on the potential influence and prognostic value of immunological processes on DCIS progression, as well as the evaluation of TILs in DCIS. Further characterisation of the immune milieu of DCIS is recommended to better understand the immune response in DCIS progression and recurrence.
Collapse
|
63
|
Loss of SDC1 Expression Is Associated with Poor Prognosis of Colorectal Cancer Patients in Northern China. DISEASE MARKERS 2019; 2019:3768708. [PMID: 31182980 PMCID: PMC6515153 DOI: 10.1155/2019/3768708] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/05/2019] [Accepted: 01/31/2019] [Indexed: 02/07/2023]
Abstract
Background Syndecan-1 (SDC1/CD138) is a key cell surface adhesion molecule essential for maintaining cell morphology and the interactions with the surrounding microenvironment. SDC1 tumor immunoexpression may be increased or decreased in epithelial malignant neoplasms compared to that in adjacent non-neoplastic tissue, depending on the type of carcinoma, and it has been correlated with various clinicopathological parameters and patient prognosis. SDC1 expression is decreased in colorectal cancer (CRC) tissue, but the relationship between prognosis and SDC1 expression in CRC patients is controversial. Methods In this study, SDC1 expression was detected in 65 adjacent non-neoplastic colorectal tissues, 477 CRCs, and 79 metastatic lymph nodes using tissue microarray. Results The data show that SDC1 decreased in CRC tissues (p ≤ 0.001) and metastatic lymph node tissues (p ≤ 0.001) compared to that in adjacent non-neoplastic colorectal tissues. Loss of SDC1 protein expression is associated with poor overall (p < 0.0001) and disease-free survival (p < 0.0001), differentiation (p = 0.017), stage (p ≤ 0.001), and lymph node metastasis (p ≤ 0.001) in CRC patients. Conclusions These data suggest that the loss of SDC1 plays an important role in CRC malignant progression. Loss of SDC1 expression indicates poor prognosis in patients from northern China with CRC.
Collapse
|
64
|
Khodadadi L, Cheng Q, Radbruch A, Hiepe F. The Maintenance of Memory Plasma Cells. Front Immunol 2019; 10:721. [PMID: 31024553 PMCID: PMC6464033 DOI: 10.3389/fimmu.2019.00721] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/18/2019] [Indexed: 12/20/2022] Open
Abstract
It is now well accepted that plasma cells can become long-lived (memory) plasma cells and secrete antibodies for months, years or a lifetime. However, the mechanisms involved in this process of humoral memory, which is crucial for both protective immunity and autoimmunity, still are not fully understood. This article will address a number of open questions. For example: Is longevity of plasma cells due to their intrinsic competence, extrinsic factors, or a combination of both? Which internal signals are involved in this process? What factors provide external support? What survival factors play a part in inflammation and autoreactive disease? Internal and external factors that contribute to the maintenance of memory long-lived plasma cells will be discussed. The aim is to provide useful additional information about the maintenance of protective and autoreactive memory plasma cells that will help researchers design effective vaccines for the induction of life-long protection against infectious diseases and to efficiently target pathogenic memory plasma cells.
Collapse
Affiliation(s)
- Laleh Khodadadi
- Deutsches Rheuma-Forschungszentrum Berlin-A Leibniz Institute, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Charité Mitte, Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Berlin, Germany
| | - Qingyu Cheng
- Deutsches Rheuma-Forschungszentrum Berlin-A Leibniz Institute, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Charité Mitte, Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Berlin, Germany
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum Berlin-A Leibniz Institute, Berlin, Germany
| | - Falk Hiepe
- Deutsches Rheuma-Forschungszentrum Berlin-A Leibniz Institute, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Charité Mitte, Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Berlin, Germany
| |
Collapse
|
65
|
Qiu H, Qi P, Liu J, Yang Y, Tan X, Xiao Y, Maitz MF, Huang N, Yang Z. Biomimetic engineering endothelium-like coating on cardiovascular stent through heparin and nitric oxide-generating compound synergistic modification strategy. Biomaterials 2019; 207:10-22. [PMID: 30947118 DOI: 10.1016/j.biomaterials.2019.03.033] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/15/2019] [Accepted: 03/22/2019] [Indexed: 01/23/2023]
Abstract
Co-immobilization of two or more molecules with different and complementary functions to prevent thrombosis, suppress smooth muscle cell (SMC) proliferation, and support endothelial cell (EC) growth is generally considered to be promising for the re-endothelialization on cardiovascular stents. However, integration of molecules with distinct therapeutic effects does not necessarily result in synergistic physiological functions due to the lack of interactions among them, limiting their practical efficacy. Herein, we apply heparin and nitric oxide (NO), two key molecules of the physiological functions of endothelium, to develop an endothelium-mimetic coating. Such coating is achieved by sequential conjugation of heparin and the NO-generating compound selenocystamine (SeCA) on an amine-bearing film of plasma polymerized allylamine. The resulting surface combines the anti-coagulant (anti-FXa) function provided by the heparin and the anti-platelet activity of the catalytically produced NO. It also endows the stents with the ability to simultaneously up-regulate α-smooth muscle actin (α-SMA) expression and to increase cyclic guanylate monophosphate (cGMP) synthesis of SMC, thereby significantly promoting their contractile phenotype and suppressing their proliferation. Importantly, this endothelium-biomimetic coating creates a favorable microenvironment for EC over SMC. These features impressively improve the antithrombogenicity, re-endothelialization and anti-restenosis of vascular stents in vivo.
Collapse
Affiliation(s)
- Hua Qiu
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Pengkai Qi
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jingxia Liu
- Physical Education Department, Southwest Jiaotong University, Chengdu, 610031, China
| | - Ying Yang
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 4059, Australia
| | - Xing Tan
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yu Xiao
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Manfred F Maitz
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden, 01069, Germany
| | - Nan Huang
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Zhilu Yang
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
66
|
Ughy B, Schmidthoffer I, Szilak L. Heparan sulfate proteoglycan (HSPG) can take part in cell division: inside and outside. Cell Mol Life Sci 2019; 76:865-871. [PMID: 30465083 PMCID: PMC11105504 DOI: 10.1007/s00018-018-2964-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 11/26/2022]
Abstract
Prior to the cytokinesis, the cell-matrix interactions should be disrupted, and the mitotic cells round up. Prerequisite of mitosis, the centrosomes duplicate, spindle fibers are generated and move away from each other to opposite sides of the cells marking the cell poles. Later, an invagination in the plasma membrane is formed a few minutes after anaphase. This furrow ingression is driven by a contractile actomyosin ring, whose assembly is regulated by RhoA GTPase. At the completion of cytokinesis, the two daughter cells are still connected by a thin intercellular bridge, which is subjected to abscission, as the terminal step of cytokinesis. Here, it is overviewed, how syndecan-4, a transmembrane, heparan sulfate proteoglycan, can contribute to these processes in a phosphorylation-dependent manner.
Collapse
Affiliation(s)
- Bettina Ughy
- Institute of Plant Biology, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, 6726, Hungary
| | - Ildiko Schmidthoffer
- Antal Wittmann Crop, Animal and Food Sciences Multidisciplinary Doctoral School, Mosonmagyaróvár, 9200, Hungary
| | - Laszlo Szilak
- Szilak Laboratories Bioinformatics and Molecule-Design Ltd., Szeged, 6723, Hungary.
- Cereal Research Non-profit Ltd., Szeged, 6726, Hungary.
| |
Collapse
|
67
|
Li Q, Ng HQ, Kang C. Secondary structure and topology of the transmembrane domain of Syndecan‐2 in detergent micelles. FEBS Lett 2019; 593:554-561. [DOI: 10.1002/1873-3468.13335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/03/2019] [Accepted: 02/07/2019] [Indexed: 01/18/2023]
Affiliation(s)
- Qingxin Li
- Institute of Chemical and Engineering Sciences Agency for Science, Technology and Research (A*STAR) Jurong Island Singapore
| | - Hui Qi Ng
- Experimental Therapeutics Centre Experimental Drug Development Centre (EDDC) Agency for Science, Technology and Research (A*STAR) Singapore Singapore
| | - CongBao Kang
- Experimental Therapeutics Centre Experimental Drug Development Centre (EDDC) Agency for Science, Technology and Research (A*STAR) Singapore Singapore
| |
Collapse
|
68
|
Letoha T, Hudák A, Kusz E, Pettkó-Szandtner A, Domonkos I, Jósvay K, Hofmann-Apitius M, Szilák L. Contribution of syndecans to cellular internalization and fibrillation of amyloid-β(1-42). Sci Rep 2019; 9:1393. [PMID: 30718543 PMCID: PMC6362000 DOI: 10.1038/s41598-018-37476-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022] Open
Abstract
Intraneuronal accumulation of amyloid-β(1-42) (Aβ1-42) is one of the earliest signs of Alzheimer's disease (AD). Cell surface heparan sulfate proteoglycans (HSPGs) have profound influence on the cellular uptake of Aβ1-42 by mediating its attachment and subsequent internalization into the cells. Colocalization of amyloid plaques with members of the syndecan family of HSPGs, along with the increased expression of syndecan-3 and -4 have already been reported in postmortem AD brains. Considering the growing evidence on the involvement of syndecans in the pathogenesis of AD, we analyzed the contribution of syndecans to cellular uptake and fibrillation of Aβ1-42. Among syndecans, the neuron specific syndecan-3 isoform increased cellular uptake of Aβ1-42 the most. Kinetics of Aβ1-42 uptake also proved to be fairly different among SDC family members: syndecan-3 increased Aβ1-42 uptake from the earliest time points, while other syndecans facilitated Aβ1-42 internalization at a slower pace. Internalized Aβ1-42 colocalized with syndecans and flotillins, highlighting the role of lipid-rafts in syndecan-mediated uptake. Syndecan-3 and 4 also triggered fibrillation of Aβ1-42, further emphasizing the pathophysiological relevance of syndecans in plaque formation. Overall our data highlight syndecans, especially the neuron-specific syndecan-3 isoform, as important players in amyloid pathology and show that syndecans, regardless of cell type, facilitate key molecular events in neurodegeneration.
Collapse
Affiliation(s)
| | | | | | | | - Ildikó Domonkos
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Katalin Jósvay
- Biological Research Centre of the Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Martin Hofmann-Apitius
- Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, 53754, Germany
| | - László Szilák
- Szilak Laboratories, Bioinformatics and Molecule-Design, Szeged, H-6723, Hungary
| |
Collapse
|
69
|
Koch J, Idzerda NMA, Dam W, Assa S, Franssen CFM, van den Born J. Plasma syndecan-1 in hemodialysis patients associates with survival and lower markers of volume status. Am J Physiol Renal Physiol 2019; 316:F121-F127. [DOI: 10.1152/ajprenal.00252.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Syndecan-1, a transmembrane heparan sulfate proteoglycan, associates with renal and cardiovascular functioning. We earlier reported syndecan-1 to be involved in renal tubular regeneration. We now examined plasma values of syndecan-1 in a hemodialysis cohort and its association with volume and inflammatory and endothelial markers in addition to outcome. Eighty-four prevalent hemodialysis patients were evaluated for their plasma syndecan-1 levels by ELISA before the start of hemodialysis, as well as 60, 180, and 240 min after start of dialysis. Patients were divided into sex-stratified tertiles based on predialysis plasma syndecan-1 levels. We studied the association between plasma levels of syndecan-1 and volume, inflammation, and endothelial markers and its association with cardiovascular events and all-cause mortality using Kaplan-Meier curves and Cox regression analyses with adjustments for gender, age, diabetes, and dialysis vintage. Predialysis syndecan-1 levels were twofold higher in men compared with women ( P = 0.0003). Patients in the highest predialysis plasma syndecan-1 tertile had a significantly higher ultrafiltration rate ( P = 0.034) and lower plasma values of BNP ( P = 0.019), pro-ANP ( P = 0.024), and endothelin ( P < 0.0001) compared with the two lower predialysis syndecan-1 tertiles. No significant associations with inflammatory markers were found. Cox regression analysis showed that patients in the highest syndecan-1 tertile had significantly less cardiovascular events and better survival compared with the lowest syndecan-1 tertile ( P = 0.02 and P = 0.005, respectively). In hemodialysis patients, higher plasma syndecan-1 levels were associated with lower concentrations of BNP, pro-ANP, and endothelin and with better patient survival. This may suggest that control of volume status in hemodialysis patients allows an adaptive tissue regenerative response as reflected by higher plasma syndecan-1 levels.
Collapse
Affiliation(s)
- Josephine Koch
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nienke M. A. Idzerda
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wendy Dam
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Solmaz Assa
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Casper F. M. Franssen
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
70
|
Christensen G, Herum KM, Lunde IG. Sweet, yet underappreciated: Proteoglycans and extracellular matrix remodeling in heart disease. Matrix Biol 2019; 75-76:286-299. [DOI: 10.1016/j.matbio.2018.01.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/22/2017] [Accepted: 01/01/2018] [Indexed: 12/20/2022]
|
71
|
Post A, Wang E, Cosgriff-Hernandez E. A Review of Integrin-Mediated Endothelial Cell Phenotype in the Design of Cardiovascular Devices. Ann Biomed Eng 2018; 47:366-380. [PMID: 30488311 DOI: 10.1007/s10439-018-02171-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022]
Abstract
Sustained biomaterial thromboresistance has long been a goal and challenge in blood-contacting device design. Endothelialization is one of the most successful strategies to achieve long-term thromboresistance of blood-contacting devices, with the endothelial cell layer providing dynamic hemostatic regulation. It is well established that endothelial cell behavior is influenced by interactions with the underlying extracellular matrix (ECM). Numerous researchers have sought to exploit these interactions to generate improved blood-contacting devices by investigating the expression of hemostatic regulators in endothelial cells on various ECM coatings. The ability to select substrates that promote endothelial cell-mediated thromboresistance is crucial to advancing material design strategies to improve cardiovascular device outcomes. This review provides an overview of endothelial cell regulation of hemostasis, the major components found within the cardiovascular basal lamina, and the interactions of endothelial cells with prominent ECM components of the basement membrane. A summary of ECM-mimetic strategies used in cardiovascular devices is provided with a focus on the effects of key adhesion modalities on endothelial cell regulators of hemostasis.
Collapse
Affiliation(s)
- Allison Post
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Ellen Wang
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Elizabeth Cosgriff-Hernandez
- Department of Biomedical Engineering, University of Texas, 107 W. Dean Keaton, BME 3.503D, 1 University Station, C0800, Austin, TX, 78712, USA.
| |
Collapse
|
72
|
Kim JM, Lee K, Kim MY, Shin HI, Jeong D. Suppressive effect of syndecan ectodomains and N-desulfated heparins on osteoclastogenesis via direct binding to macrophage-colony stimulating factor. Cell Death Dis 2018; 9:1119. [PMID: 30389911 PMCID: PMC6215006 DOI: 10.1038/s41419-018-1167-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/20/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022]
Abstract
Syndecans, a family of cell surface heparan sulfate proteoglycans, regulate cell differentiation via binding of their heparan sulfate chains to growth factors and cytokines and play a role in tumor growth and progression, wound repair, and intestinal mucosal damage. However, the functional and mechanistic roles of syndecans in osteoclast differentiation and bone metabolism are yet unclear. Here, we demonstrated that post-translationally glycosylated ectodomains of syndecan-1 to 4 obtained from mammalian cells efficiently suppressed osteoclast differentiation compared to those obtained from Escherichia coli with no systems for glycosylation. A concomitant decrease in the expression of osteoclast markers such as nuclear factor of activated T cells 1 (NFATc1), c-Fos, and ATP6V0D2 was observed. In addition, heparan sulfate and selectively N-desulfated heparin derivatives with 2-O- and 6-O-sulfate groups and no anticoagulant activity in blood inhibited osteoclast differentiation. The inhibitory effects of syndecan ectodomains, heparan sulfate, and N-desulfated heparin derivatives on osteoclast differentiation were attributed to their direct binding to the macrophage-colony stimulating factor (M-CSF), resulting in the blocking of M-CSF-mediated downstream signals such as extracellular signal-regulated kinase (ERK), c-JUN N-terminal kinase (JNK), p38, and Akt. Furthermore, mice injected with syndecan ectodomains, heparan sulfate, and N-desulfated heparin derivatives into periosteal regions of calvaria showed reduction in the formation of tartrate-resistant acid phosphatase (TRAP)-positive mature osteoclasts on the calvarial bone surface, thereby exhibiting decreased bone resorption. Together, these results revealed a novel role of heparan sulfate chains of syndecan ectodomains in the regulation of osteoclast differentiation.
Collapse
Affiliation(s)
- Jin-Man Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, 42415, Korea.,Asan Medical Center, Asan Institute for Life Sciences, Seoul, 26493, Korea
| | - Kyunghee Lee
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Mi Yeong Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Hong-In Shin
- Department of Oral Pathology, Institute for Hard Tissue and Bio-Tooth Regeneration, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Daewon Jeong
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, 42415, Korea.
| |
Collapse
|
73
|
Song SH, Jang WJ, Hwang J, Park B, Jang JH, Seo YH, Yang CH, Lee S, Jeong CH. Transcriptome profiling of whisker follicles in methamphetamine self-administered rats. Sci Rep 2018; 8:11420. [PMID: 30061674 PMCID: PMC6065325 DOI: 10.1038/s41598-018-29772-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/18/2018] [Indexed: 12/12/2022] Open
Abstract
Methamphetamine (MA) is a highly addictive psychostimulant that disturbs the central nervous system; therefore, diagnosis of MA addiction is important in clinical and forensic toxicology. In this study, a MA self-administration rat model was used to illustrate the gene expression profiling of the rewarding effect caused by MA. RNA-sequencing was performed to examine changes in gene expression in rat whisker follicles collected before self-administration, after MA self-administration, and after withdrawal sessions. We identified six distinct groups of genes, with statistically significant expression patterns. By constructing the functional association network of these genes and performing the subsequent topological analysis, we identified 43 genes, which have the potential to regulate MA reward and addiction. The gene pathways were then analysed using the Reactome and Knowledgebase for Addiction-Related Gene database, and it was found that genes and pathways associated with Alzheimer's disease and the heparan sulfate biosynthesis were enriched in MA self-administration rats. The findings suggest that changes of the genes identified in rat whisker follicles may be useful indicators of the rewarding effect of MA. Further studies are needed to provide a comprehensive understanding of MA addiction.
Collapse
Affiliation(s)
- Sang-Hoon Song
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Won-Jun Jang
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jihye Hwang
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jung-Hee Jang
- School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Young-Ho Seo
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Chae Ha Yang
- College of Oriental Medicine, Daegu Hanny University, Daegu, 42158, Republic of Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
74
|
Kero D, Bilandzija TS, Arapovic LL, Vukojevic K, Saraga-Babic M. Syndecans and Enzymes Involved in Heparan Sulfate Biosynthesis and Degradation Are Differentially Expressed During Human Odontogenesis. Front Physiol 2018; 9:732. [PMID: 29962964 PMCID: PMC6010574 DOI: 10.3389/fphys.2018.00732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/25/2018] [Indexed: 01/01/2023] Open
Abstract
Syndecans belong to a four-member family of cell surface heparan sulfate proteoglycans (HSPGs) abundantly present in various tissues. They are primarily recognized as extracellular matrix (ECM) receptors able to bind various ECM components and form gradients of morphogens and growth factors. Syndecans are composed of core protein with distinctive cytoplasmic, transmembrane, and extracellular domains to which several HS glycosaminoglycan (GAG) chains are covalently attached. In development of composite organs, such as teeth, expression patterns of syndecans display temporo-spatial shifts between epithelial and mesenchymal tissue compartments. Along with diverse functional properties of syndecans and generally large number of their interactors due to HS GAG chain content, this suggests possible involvement of syndecans in modulation of epithelial-to-mesenchymal crosstalk. Functional versatility of syndecans greatly depends upon the biochemical properties of attached HS GAG chains. These are specifically determined during the HS biosynthesis by the combinatorial action of glycosyl-transferases (Exts/EXTs) and bi-functional sulfotransferases (Ndsts/NDSTs), as well as by post-biosynthetic enzymatic cleavage of HS by the only active endoglucuronidase in mammals, heparanase 1 (Hpse1/HPSE1). Matching the essential requirement for HS during organogenesis, null-mutant animals for genes encoding these enzymes display severe developmental anomalies of mineralized tissues (including teeth) with embryonic or perinatal lethality. In this study, we analyzed expression of syndecan HSPGs (syndecans 1, 2, and 4), enzymes involved in HS biosynthesis (EXT1, NDST1, NDST2) and HS cleavage (HPSE1) in human tooth germs during the early stages of odontogenesis. All of the investigated factors displayed temporo-spatial differences in expression patterns, and some of them showed distinctive asymmetries of expression domains. Our findings suggest that these factors might be differentially involved in cellular processes which take place during the early odontogenic sequence in humans.
Collapse
Affiliation(s)
- Darko Kero
- Department of Dental Morphology and Anthropology, Study Program of Dental Medicine, School of Medicine, University of Split, Split, Croatia
| | - Tanja Simic Bilandzija
- Department of Maxillofacial Surgery, University Clinical Hospital Mostar, Mostar, Bosnia and Herzegovina.,Study Program of Dental Medicine, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Lidija Lasic Arapovic
- Study Program of Dental Medicine, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina.,Primary Health Care Center Mostar, Mostar, Bosnia and Herzegovina
| | - Katarina Vukojevic
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Split, Croatia
| | - Mirna Saraga-Babic
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Split, Croatia
| |
Collapse
|
75
|
Functional relevance of in vivo half antibody exchange of an IgG4 therapeutic antibody-drug conjugate. PLoS One 2018; 13:e0195823. [PMID: 29672587 PMCID: PMC5908158 DOI: 10.1371/journal.pone.0195823] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 03/30/2018] [Indexed: 11/22/2022] Open
Abstract
An increasing number of monoclonal antibodies and derivatives such as antibody-drug conjugates (ADC) are of the IgG1 and IgG4 isotype with distinct structural and functional properties. In cases where antibody-mediated cytotoxicity is not desired, IgG4 is often used, as its Fc region is relatively poor at inducing antibody-dependent cell-mediated or complement-dependent cytotoxicity. IgG4 ADCs with highly cytotoxic drugs against proliferating target cells but which lack or have diminished antibody effector functions against quiescent cells may have a favorable safety profile compared to IgG1. Another unique property of the IgG4 subclass is the capability to exchange half antibodies in vivo creating randomly bispecific antibodies. To investigate the functional properties of process-derived antibody species, and determine the influence of shuffling on the therapeutic efficacy, several model antibodies on the basis of the anti-CD138 antibody-drug conjugate BT062 (Indatuximab ravtansine) were generated: (I) A wild type nBT062, (II) a stable nBT062 comprising mutations to prevent half-antibody exchange, (III) a half nBT062 lacking covalent binding between two heavy chains and (IV) a stabilized, bispecific nBT062-natalizumab antibody with a second, monovalent specificity against CD49d. All nBT062 model variants were capable of CD138-specific binding and antigen-mediated internalization into cells. Furthermore, all nBT062 models inhibited tumor growth in vitro after conjugation with the maytansinoid DM4. The in vivo effects of the different molecular variants were assessed in the MAXF1322 xenograft model. The bispecific nBT062-natalizumab-DM4 demonstrated the least efficacy and was only moderately active even without the co-administration of a human IgG preparation. Wild type, stable and half nBT062-DM4 models demonstrated great anti-tumor activities. The efficacy of wild type and half nBT062-DM4 was reduced in the presence of IgG, while stable nBT062-DM4 was only marginally influenced. These pre-clinical data demonstrate the advantage of introducing half-antibody exchange-preventing mutations into therapeutic IgG4-based antibody drug-conjugates.
Collapse
|
76
|
Abstract
Syndecan-1 (Sdc1) is a major cell surface heparan sulfate (HS) proteoglycan of epithelial cells, a cell type targeted by many bacterial pathogens early in their pathogenesis. Loss of Sdc1 in mice is a gain-of-function mutation that significantly decreases the susceptibility to several bacterial infections, suggesting that subversion of Sdc1 is an important virulence strategy. HS glycosaminoglycan (GAG) chains of cell surface Sdc1 promote bacterial pathogenesis by facilitating the attachment of bacteria to host cells. Engagement of cell surface Sdc1 HS chains by bacterial adhesins transmits signal through the highly conserved Sdc1 cytoplasmic domain, which can lead to uptake of intracellular bacterial pathogens. On the other hand, several bacteria that do not require Sdc1 for their attachment and invasion stimulate Sdc1 shedding and exploit the capacity of Sdc1 ectodomain HS GAGs to disarm innate defense mechanisms to evade immune clearance. Recent data suggest that select HS sulfate motifs, and not the overall charge of HS, are important in the inhibition of innate immune mechanisms. Here, we discuss several examples of Sdc1 subversion in bacterial infections.
Collapse
|
77
|
Tsoyi K, Chu SG, Patino-Jaramillo NG, Wilder J, Villalba J, Doyle-Eisele M, McDonald J, Liu X, El-Chemaly S, Perrella MA, Rosas IO. Syndecan-2 Attenuates Radiation-induced Pulmonary Fibrosis and Inhibits Fibroblast Activation by Regulating PI3K/Akt/ROCK Pathway via CD148. Am J Respir Cell Mol Biol 2018; 58:208-215. [PMID: 28886261 DOI: 10.1165/rcmb.2017-0088oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Radiation-induced pulmonary fibrosis is a severe complication of patients treated with thoracic irradiation. We have previously shown that syndecan-2 reduces fibrosis by exerting alveolar epithelial cytoprotective effects. Here, we investigate whether syndecan-2 attenuates radiation-induced pulmonary fibrosis by inhibiting fibroblast activation. C57BL/6 wild-type mice and transgenic mice that overexpress human syndecan-2 in alveolar macrophages were exposed to 14 Gy whole-thoracic radiation. At 24 weeks after irradiation, lungs were collected for histological, protein, and mRNA evaluation of pulmonary fibrosis, profibrotic gene expression, and α-smooth muscle actin (α-SMA) expression. Mouse lung fibroblasts were activated with transforming growth factor (TGF)-β1 in the presence or absence of syndecan-2. Cell proliferation, migration, and gel contraction were assessed at different time points. Irradiation resulted in significantly increased mortality and pulmonary fibrosis in wild-type mice that was associated with elevated lung expression of TGF-β1 downstream target genes and cell death compared with irradiated syndecan-2 transgenic mice. In mouse lung fibroblasts, syndecan-2 inhibited α-SMA expression, cell contraction, proliferation, and migration induced by TGF-β1. Syndecan-2 attenuated phosphoinositide 3-kinase/serine/threonine kinase/Rho-associated coiled-coil kinase signaling and serum response factor binding to the α-SMA promoter. Syndecan-2 attenuates pulmonary fibrosis in mice exposed to radiation and inhibits TGF-β1-induced fibroblast-myofibroblast differentiation, migration, and proliferation by down-regulating phosphoinositide 3-kinase/serine/threonine kinase/Rho-associated coiled-coil kinase signaling and blocking serum response factor binding to the α-SMA promoter via CD148. These findings suggest that syndecan-2 has potential as an antifibrotic therapy in radiation-induced lung fibrosis.
Collapse
Affiliation(s)
- Konstantin Tsoyi
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Sarah G Chu
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | | | - Julie Wilder
- 2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Julian Villalba
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and.,2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Melanie Doyle-Eisele
- 2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Jacob McDonald
- 2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Xiaoli Liu
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Souheil El-Chemaly
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Mark A Perrella
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Ivan O Rosas
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and.,2 Pulmonary Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| |
Collapse
|
78
|
RNA-Seq reveals seven promising candidate genes affecting the proportion of thick egg albumen in layer-type chickens. Sci Rep 2017; 7:18083. [PMID: 29273734 PMCID: PMC5741707 DOI: 10.1038/s41598-017-18389-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/11/2017] [Indexed: 01/07/2023] Open
Abstract
Eggs with a much higher proportion of thick albumen are preferred in the layer industry, as they are favoured by consumers. However, the genetic factors affecting the thick egg albumen trait have not been elucidated. Using RNA sequencing, we explored the magnum transcriptome in 9 Rhode Island white layers: four layers with phenotypes of extremely high ratios of thick to thin albumen (high thick albumen, HTA) and five with extremely low ratios (low thick albumen, LTA). A total of 220 genes were differentially expressed, among which 150 genes were up-regulated and 70 were down-regulated in the HTA group compared with the LTA group. Gene Ontology (GO) analysis revealed that the up-regulated genes in HTA were mainly involved in a wide range of regulatory functions. In addition, a large number of these genes were related to glycosphingolipid biosynthesis, focal adhesion, ECM-receptor interactions and cytokine-cytokine receptor interactions. Based on functional analysis, ST3GAL4, FUT4, ITGA2, SDC3, PRLR, CDH4 and GALNT9 were identified as promising candidate genes for thick albumen synthesis and metabolism during egg formation. These results provide new insights into the molecular mechanisms of egg albumen traits and may contribute to future breeding strategies that optimise the proportion of thick egg albumen.
Collapse
|
79
|
Abstract
Syndecans comprise a major family of cell surface heparan sulfate proteoglycans (HSPGs). Syndecans are composed of sulfated glycosaminoglycans (GAGs), heparan sulfate (HS) or both HS and chondroitin sulfate (CS), attached covalently to core proteins. Syndecans regulate many cellular processes, such as adhesion, proliferation, and migration. Syndecans bind and regulate molecules primarily through their HS chains, but do not bind to all HS/heparin-binding molecules. Furthermore, mice ablated for the syndecan-1 or -4 gene do not show major developmental abnormalities, but they do show striking pathological phenotypes when challenged with infectious or inflammatory stimuli and conditions, suggesting that certain functions of syndecans are specific and cannot be compensated for by other syndecans or other HSPGs. These observations underscore the physiological importance of syndecans and indicate a need to study the activities of isolated native syndecans to define their molecular and cellular functions, and to establish their biological significance. Here we describe methods to isolate syndecans and several assays to analyze their functions.
Collapse
Affiliation(s)
- Pyong Woo Park
- Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
80
|
Papy-Garcia D, Albanese P. Heparan sulfate proteoglycans as key regulators of the mesenchymal niche of hematopoietic stem cells. Glycoconj J 2017; 34:377-391. [PMID: 28577070 DOI: 10.1007/s10719-017-9773-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 12/21/2022]
Abstract
The complex microenvironment that surrounds hematopoietic stem cells (HSCs) in the bone marrow niche involves different coordinated signaling pathways. The stem cells establish permanent interactions with distinct cell types such as mesenchymal stromal cells, osteoblasts, osteoclasts or endothelial cells and with secreted regulators such as growth factors, cytokines, chemokines and their receptors. These interactions are mediated through adhesion to extracellular matrix compounds also. All these signaling pathways are important for stem cell fates such as self-renewal, proliferation or differentiation, homing and mobilization, as well as for remodeling of the niche. Among these complex molecular cues, this review focuses on heparan sulfate (HS) structures and functions and on the role of enzymes involved in their biosynthesis and turnover. HS associated to core protein, constitute the superfamily of heparan sulfate proteoglycans (HSPGs) present on the cell surface and in the extracellular matrix of all tissues. The key regulatory effects of major medullar HSPGs are described, focusing on their roles in the interactions between hematopoietic stem cells and their endosteal niche, and on their ability to interact with Heparin Binding Proteins (HBPs). Finally, according to the relevance of HS moieties effects on this complex medullar niche, we describe recent data that identify HS mimetics or sulfated HS signatures as new glycanic tools and targets, respectively, for hematopoietic and mesenchymal stem cell based therapeutic applications.
Collapse
Affiliation(s)
- Dulce Papy-Garcia
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France
| | - Patricia Albanese
- CRRET Laboratory, Université Paris Est, EA 4397 Université Paris Est Créteil, ERL CNRS 9215, F-94010, Créteil, France.
| |
Collapse
|
81
|
Miligy I, Mohan P, Gaber A, Aleskandarany MA, Nolan CC, Diez-Rodriguez M, Mukherjee A, Chapman C, Ellis IO, Green AR, Rakha EA. Prognostic significance of tumour infiltrating B lymphocytes in breast ductal carcinoma in situ. Histopathology 2017; 71:258-268. [PMID: 28326600 DOI: 10.1111/his.13217] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/18/2017] [Indexed: 12/21/2022]
Abstract
AIMS Tumour-infiltrating lymphocytes (TILs) are an important component of the immune response to cancer and have a prognostic value in breast cancer. Although several studies have investigated the role of T lymphocytes in breast cancer, the role of B lymphocytes (TIL-Bs) in ductal carcinoma in situ (DCIS) remains uncertain. This study aimed to assess the role of TIL-Bs in DCIS. METHODS AND RESULTS Eighty DCIS cases (36 pure DCIS and 44 mixed with invasive cancer) were stained immunohistochemically for B lineage markers CD19, CD20 and the plasma cell marker CD138. TIL-Bs density and localization were assessed, including relation to the in-situ and invasive components. An association with clinicopathological data and patient outcome was performed. Pure DCIS showed a higher number of TIL-Bs and lymphoid aggregates than DCIS associated with invasion. In pure DCIS, a higher number of peri- and paratumoral TIL-Bs was associated significantly with large tumour size (P = 0.016), hormone receptor (ER/PR) negative (P = 0.008) and HER2+ status (P = 0.010). In tumours with mixed DCIS and invasive components, cases with high-density B lymphocytes, irrespective of their location or topographic distribution, were associated significantly with variables of poor prognosis, including larger size, high grade, lymphovascular invasion, lymph node metastases, ER/PR-negative and HER2+ status. Outcome analysis showed that pure DCIS associated with higher numbers of B lymphocytes had shorter recurrence-free interval (P = 0.04); however, the association was not significant with the CD138+ plasma cell count (P = 0.07). CONCLUSION Assessment of TIL-B cells based on location and topographic distribution can provide prognostic information. Validation in a larger cohort is warranted.
Collapse
Affiliation(s)
- Islam Miligy
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology Department, Faculty of Medicine, Monofiya University, Egypt
| | - Priya Mohan
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Ahmed Gaber
- General Surgery Department, Faculty of Medicine, Monofiya University, Egypt
| | - Mohammed A Aleskandarany
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology Department, Faculty of Medicine, Monofiya University, Egypt
| | - Christopher C Nolan
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Maria Diez-Rodriguez
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Abhik Mukherjee
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Caroline Chapman
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Ian O Ellis
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R Green
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology Department, Faculty of Medicine, Monofiya University, Egypt
| |
Collapse
|
82
|
Guan J, Bywaters SM, Brendle SA, Ashley RE, Makhov AM, Conway JF, Christensen ND, Hafenstein S. Cryoelectron Microscopy Maps of Human Papillomavirus 16 Reveal L2 Densities and Heparin Binding Site. Structure 2017; 25:253-263. [PMID: 28065506 DOI: 10.1016/j.str.2016.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/07/2016] [Accepted: 12/12/2016] [Indexed: 11/24/2022]
Abstract
Human papillomavirus (HPV) is a significant health burden and leading cause of virus-induced cancers. The current commercial vaccines are genotype specific and provide little therapeutic benefit to patients with existing HPV infections. Host entry mechanisms represent an excellent target for alternative therapeutics, but HPV receptor use, the details of cell attachment, and host entry are inadequately understood. Here we present near-atomic resolution structures of the HPV16 capsid and HPV16 in complex with heparin, both determined from cryoelectron micrographs collected with direct electron detection technology. The structures clarify details of capsid architecture for the first time, including variation in L1 major capsid protein conformation and putative location of L2 minor protein. Heparin binds specifically around the capsid icosahedral vertices and may recapitulate the earliest stage of infection, providing a framework for continuing biochemical, genetic, and biophysical studies.
Collapse
Affiliation(s)
- Jian Guan
- Division of Infectious Diseases, Department of Medicine, Penn State College of Medicine, The Pennsylvania State University College of Medicine, Mail Code H036, 500 University Drive, P.O. Box 850, Hershey, PA 17033-0850, USA
| | - Stephanie M Bywaters
- Department of Pathology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Sarah A Brendle
- Department of Pathology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Robert E Ashley
- Division of Infectious Diseases, Department of Medicine, Penn State College of Medicine, The Pennsylvania State University College of Medicine, Mail Code H036, 500 University Drive, P.O. Box 850, Hershey, PA 17033-0850, USA
| | - Alexander M Makhov
- Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 5th Avenue, Pittsburgh, PA 15260, USA
| | - James F Conway
- Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 5th Avenue, Pittsburgh, PA 15260, USA
| | - Neil D Christensen
- Department of Pathology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Susan Hafenstein
- Division of Infectious Diseases, Department of Medicine, Penn State College of Medicine, The Pennsylvania State University College of Medicine, Mail Code H036, 500 University Drive, P.O. Box 850, Hershey, PA 17033-0850, USA.
| |
Collapse
|
83
|
Huang YZ, Wang Q, Zhang CL, Fang XT, Song EL, Chen H. Genetic Variants in SDC3 Gene are Significantly Associated with Growth Traits in Two Chinese Beef Cattle Breeds. Anim Biotechnol 2016; 27:190-8. [PMID: 27119984 DOI: 10.1080/10495398.2016.1164178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Identification of the genes and polymorphisms underlying quantitative traits, and understanding these genes and polymorphisms affect economic growth traits, are important for successful marker-assisted selection and more efficient management strategies in commercial cattle (Bos taurus) population. Syndecan-3 (SDC3), a member of the syndecan family of type I transmembrane heparan sulfate proteoglycans is a novel regulator of feeding behavior and body weight. The aim of this study is to examine the association of the SDC3 polymorphism with growth traits in Chinese Jiaxian and Qinchuan cattle breeds (). Four single nucleotide polymorphisms (SNPs: 1-4) were detected in 555 cows from three Chinese native cattle breeds by means of sequencing pooled DNA samples and polymerase chain reaction-single stranded conformational polymorphism (PCR-SSCP) methods. We found one SNP (g.28362A > G) in intron and three SNPs (g.30742T > G, g.30821C > T and 33418 A > G) in exons. The statistical analyses indicated that these SNPs of SDC3 gene were associated with bovine body height, body length, chest circumference, and circumference of cannon bone (P < 0.05). The mutant-type variant was superior for growth traits; the heterozygote was associated with higher growth traits compared to wild-type homozygote. Our result confirms the polymorphisms in the SDC3 gene are associated with growth traits that may be used for marker-assisted selection in beef cattle breeding programs.
Collapse
Affiliation(s)
- Yong-Zhen Huang
- a Institute of Cellular and Molecular Biology , Jiangsu Normal University , Xuzhou Jiangsu , China.,b College of Animal Science and Technology , Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture , Yangling Shaanxi , China
| | - Qin Wang
- a Institute of Cellular and Molecular Biology , Jiangsu Normal University , Xuzhou Jiangsu , China
| | - Chun-Lei Zhang
- a Institute of Cellular and Molecular Biology , Jiangsu Normal University , Xuzhou Jiangsu , China
| | - Xing-Tang Fang
- a Institute of Cellular and Molecular Biology , Jiangsu Normal University , Xuzhou Jiangsu , China
| | - En-Liang Song
- c Institute of Animal Husbandry and Veterinary , Shandong Academy of Agricultural Sciences , Jinan , Shandong , China
| | - Hong Chen
- a Institute of Cellular and Molecular Biology , Jiangsu Normal University , Xuzhou Jiangsu , China
| |
Collapse
|
84
|
Discovery of an enzyme and substrate selective inhibitor of ADAM10 using an exosite-binding glycosylated substrate. Sci Rep 2016; 6:11. [PMID: 28442704 PMCID: PMC5431342 DOI: 10.1038/s41598-016-0013-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/12/2016] [Indexed: 02/01/2023] Open
Abstract
ADAM10 and ADAM17 have been shown to contribute to the acquired drug resistance of HER2-positive breast cancer in response to trastuzumab. The majority of ADAM10 and ADAM17 inhibitor development has been focused on the discovery of compounds that bind the active site zinc, however, in recent years, there has been a shift from active site to secondary substrate binding site (exosite) inhibitor discovery in order to identify non-zinc-binding molecules. In the present work a glycosylated, exosite-binding substrate of ADAM10 and ADAM17 was utilized to screen 370,276 compounds from the MLPCN collection. As a result of this uHTS effort, a selective, time-dependent, non-zinc-binding inhibitor of ADAM10 with Ki = 883 nM was discovered. This compound exhibited low cell toxicity and was able to selectively inhibit shedding of known ADAM10 substrates in several cell-based models. We hypothesize that differential glycosylation of these cognate substrates is the source of selectivity of our novel inhibitor. The data indicate that this novel inhibitor can be used as an in vitro and, potentially, in vivo, probe of ADAM10 activity. Additionally, results of the present and prior studies strongly suggest that glycosylated substrate are applicable as screening agents for discovery of selective ADAM probes and therapeutics.
Collapse
|
85
|
Malhotra SS, Banerjee P, Gupta SK. Regulation of trophoblast differentiation during embryo implantation and placentation: Implications in pregnancy complications. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jrhm.2016.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
86
|
Melleby AO, Strand ME, Romaine A, Herum KM, Skrbic B, Dahl CP, Sjaastad I, Fiane AE, Filmus J, Christensen G, Lunde IG. The Heparan Sulfate Proteoglycan Glypican-6 Is Upregulated in the Failing Heart, and Regulates Cardiomyocyte Growth through ERK1/2 Signaling. PLoS One 2016; 11:e0165079. [PMID: 27768722 PMCID: PMC5074531 DOI: 10.1371/journal.pone.0165079] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/05/2016] [Indexed: 11/18/2022] Open
Abstract
Pressure overload is a frequent cause of heart failure. Heart failure affects millions of patients worldwide and is a major cause of morbidity and mortality. Cell surface proteoglycans are emerging as molecular players in cardiac remodeling, and increased knowledge about their regulation and function is needed for improved understanding of cardiac pathogenesis. Here we investigated glypicans (GPC1-6), a family of evolutionary conserved heparan sulfate proteoglycans anchored to the extracellular leaflet of the cell membrane, in experimental and clinical heart failure, and explored the function of glypican-6 in cardiac cells in vitro. In mice subjected to pressure overload by aortic banding (AB), we observed elevated glypican-6 levels during hypertrophic remodeling and dilated, end-stage heart failure. Consistently, glypican-6 mRNA was elevated in left ventricular myocardium from explanted hearts of patients with end-stage, dilated heart failure with reduced ejection fraction. Glypican-6 levels correlated negatively with left ventricular ejection fraction in patients, and positively with lung weight after AB in mice. Glypican-6 mRNA was expressed in both cardiac fibroblasts and cardiomyocytes, and the corresponding protein displayed different sizes in the two cell types due to tissue-specific glycanation. Importantly, adenoviral overexpression of glypican-6 in cultured cardiomyocytes increased protein synthesis and induced mRNA levels of the pro-hypertrophic signature gene ACTA1 and the hypertrophy and heart failure signature genes encoding natriuretic peptides, NPPA and NPPB. Overexpression of GPC6 induced ERK1/2 phosphorylation, and co-treatment with the ERK inhibitor U0126 attenuated the GPC6-induced increase in NPPA, NPPB and protein synthesis. In conclusion, our data suggests that glypican-6 plays a role in clinical and experimental heart failure progression by regulating cardiomyocyte growth through ERK signaling.
Collapse
Affiliation(s)
- Arne O. Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- * E-mail:
| | - Mari E. Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Andreas Romaine
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Kate M. Herum
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Biljana Skrbic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Christen P. Dahl
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Arnt E. Fiane
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Jorge Filmus
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, University of Oslo, Oslo, Norway
| |
Collapse
|
87
|
Malmos KG, Bjerring M, Jessen CM, Nielsen EHT, Poulsen ET, Christiansen G, Vosegaard T, Skrydstrup T, Enghild JJ, Pedersen JS, Otzen DE. How Glycosaminoglycans Promote Fibrillation of Salmon Calcitonin. J Biol Chem 2016; 291:16849-62. [PMID: 27281819 PMCID: PMC4974396 DOI: 10.1074/jbc.m116.715466] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 06/07/2016] [Indexed: 12/27/2022] Open
Abstract
Glycosaminoglycans (GAGs) bind all known amyloid plaques and help store protein hormones in (acidic) granular vesicles, but the molecular mechanisms underlying these important effects are unclear. Here we investigate GAG interactions with the peptide hormone salmon calcitonin (sCT). GAGs induce fast sCT fibrillation at acidic pH and only bind monomeric sCT at acidic pH, inducing sCT helicity. Increasing GAG sulfation expands the pH range for binding. Heparin, the most highly sulfated GAG, binds sCT in the pH interval 3-7. Small angle x-ray scattering indicates that sCT monomers densely decorate and pack single heparin chains, possibly via hydrophobic patches on helical sCT. sCT fibrillates without GAGs, but heparin binding accelerates the process by decreasing the otherwise long fibrillation lag times at low pH and accelerates fibril growth rates at neutral pH. sCT·heparin complexes form β-sheet-rich heparin-covered fibrils. Solid-state NMR reveals that heparin does not alter the sCT fibrillary core around Lys(11) but makes changes to Val(8) on the exterior side of the β-strand, possibly through contacts to Lys(18) Thus GAGs significantly modulate sCT fibrillation in a pH-dependent manner by interacting with both monomeric and aggregated sCT.
Collapse
Affiliation(s)
- Kirsten Gade Malmos
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark, Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Morten Bjerring
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark, Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000 Aarhus C, Denmark
| | - Christian Moestrup Jessen
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Erik Holm Toustrup Nielsen
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Ebbe T Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000 Aarhus, Denmark, and
| | - Thomas Vosegaard
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark, Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000 Aarhus C, Denmark
| | - Troels Skrydstrup
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Jan Skov Pedersen
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark, Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000 Aarhus C, Denmark
| | - Daniel E Otzen
- From the Interdisciplinary Nanoscience Center (iNANO) and Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark, Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark,
| |
Collapse
|
88
|
Kühl AA, Erben U, Cieluch C, Spieckermann S, Gröne J, Lohneis P, Pape UF, Arsenic R, Utku N. Tissue-infiltrating plasma cells are an important source of carboxylesterase 2 contributing to the therapeutic efficacy of prodrugs. Cancer Lett 2016; 378:51-8. [DOI: 10.1016/j.canlet.2016.04.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 12/16/2022]
|
89
|
|
90
|
Cubela M, Soljic V, Kero D, Vukojevic K, Govorko DK, Saraga-Babic M. Comparison of proliferation, apoptosis and expression of syndecan-1 and α-SMA in edentulous ridge oral mucosa of successful and early failed submerged dental implants—An immunohistochemical study. Arch Oral Biol 2016; 66:155-64. [DOI: 10.1016/j.archoralbio.2016.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/18/2016] [Accepted: 02/26/2016] [Indexed: 11/30/2022]
|
91
|
Wang R, Wang X, Ni B, Huan CC, Wu JQ, Wen LB, Liao Y, Tong GZ, Ding C, Fan HJ, Mao X. Syndecan-4, a PRRSV attachment factor, mediates PRRSV entry through its interaction with EGFR. Biochem Biophys Res Commun 2016; 475:230-7. [PMID: 27208778 DOI: 10.1016/j.bbrc.2016.05.084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 10/21/2022]
Abstract
The causative agent of porcine reproductive and respiratory syndrome is the PRRS virus (PRRSV), an enveloped, single-stranded and positive-sense RNA virus. The host factors and mechanisms that are involved in PRRSV entry are still largely unknown. In our present studies, we found that syndecan-4, one of the heparan sulfate proteoglycans, plays a critical role in PRRSV entry, especially in PRRSV attachment. Moreover, EGFR interacts with syndecan-4 in MACR-145 cells and disruption of their interaction impaired PRRSV entry. Furthermore, EGFR inhibitor AG1478 or syndecan-4 derived peptide SSTN87-131 inhibited syndecan-4 endocytosis induced by PRRSV entry. Altogether, syndecan-4, a PRRSV attachment factor, mediated PRRSV entry by interacting with EGFR.
Collapse
Affiliation(s)
- Rui Wang
- College of Veterinary Medicine, Nanjing Agricultural University, China.
| | - Xin Wang
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Bo Ni
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Chang-Chao Huan
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Jia-Qiang Wu
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, China
| | - Li-Bin Wen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Science, China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Guang-Zhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Hong-Jie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Xiang Mao
- College of Veterinary Medicine, Nanjing Agricultural University, China; Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China.
| |
Collapse
|
92
|
Anderson EJP, Çakir I, Carrington SJ, Cone RD, Ghamari-Langroudi M, Gillyard T, Gimenez LE, Litt MJ. 60 YEARS OF POMC: Regulation of feeding and energy homeostasis by α-MSH. J Mol Endocrinol 2016; 56:T157-74. [PMID: 26939593 PMCID: PMC5027135 DOI: 10.1530/jme-16-0014] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022]
Abstract
The melanocortin peptides derived from pro-opiomelanocortin (POMC) were originally understood in terms of the biological actions of α-melanocyte-stimulating hormone (α-MSH) on pigmentation and adrenocorticotrophic hormone on adrenocortical glucocorticoid production. However, the discovery of POMC mRNA and melanocortin peptides in the CNS generated activities directed at understanding the direct biological actions of melanocortins in the brain. Ultimately, discovery of unique melanocortin receptors expressed in the CNS, the melanocortin-3 (MC3R) and melanocortin-4 (MC4R) receptors, led to the development of pharmacological tools and genetic models leading to the demonstration that the central melanocortin system plays a critical role in the regulation of energy homeostasis. Indeed, mutations in MC4R are now known to be the most common cause of early onset syndromic obesity, accounting for 2-5% of all cases. This review discusses the history of these discoveries, as well as the latest work attempting to understand the molecular and cellular basis of regulation of feeding and energy homeostasis by the predominant melanocortin peptide in the CNS, α-MSH.
Collapse
Affiliation(s)
- Erica J P Anderson
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Isin Çakir
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Sheridan J Carrington
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Roger D Cone
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Masoud Ghamari-Langroudi
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Taneisha Gillyard
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA Meharry Medical CollegeDepartment of Neuroscience and Pharmacology, Nashville, Tennessee, USA
| | - Luis E Gimenez
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Michael J Litt
- Department of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
93
|
The Cryptosporidium parvum C-Type Lectin CpClec Mediates Infection of Intestinal Epithelial Cells via Interactions with Sulfated Proteoglycans. Infect Immun 2016; 84:1593-1602. [PMID: 26975991 DOI: 10.1128/iai.01410-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 03/07/2016] [Indexed: 12/18/2022] Open
Abstract
The apicomplexan parasite Cryptosporidium causes significant diarrheal disease worldwide. Effective anticryptosporidial agents are lacking, in part because the molecular mechanisms underlying Cryptosporidium-host cell interactions are poorly understood. Previously, we identified and characterized a novel Cryptosporidium parvum C-type lectin domain-containing mucin-like glycoprotein, CpClec. In this study, we evaluated the mechanisms underlying interactions of CpClec with intestinal epithelial cells by using an Fc-tagged recombinant protein. CpClec-Fc displayed Ca(2+)-dependent, saturable binding to HCT-8 and Caco-2 cells and competitively inhibited C. parvum attachment to and infection of HCT-8 cells. Binding of CpClec-Fc was specifically inhibited by sulfated glycosaminoglycans, particularly heparin and heparan sulfate. Binding was reduced after the removal of heparan sulfate and following the inhibition of glycosaminoglycan synthesis or sulfation in HCT-8 cells. Like CpClec-Fc binding, C. parvum attachment to and infection of HCT-8 cells were inhibited by glycosaminoglycans and were reduced after heparan sulfate removal or inhibition of glycosaminoglycan synthesis or sulfation. Lastly, CpClec-Fc binding and C. parvum sporozoite attachment were significantly decreased in CHO cell mutants defective in glycosaminoglycan synthesis. Together, these results indicate that CpClec is a novel C-type lectin that mediates C. parvum attachment and infection via Ca(2+)-dependent binding to sulfated proteoglycans on intestinal epithelial cells.
Collapse
|
94
|
|
95
|
Nikolovska K, Spillmann D, Seidler DG. Uronyl 2-O sulfotransferase potentiates Fgf2-induced cell migration. J Cell Sci 2016; 128:460-71. [PMID: 25480151 DOI: 10.1242/jcs.152660] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fibroblast growth factor 2 (Fgf2) is involved in several biological functions. Fgf2 requires glycosaminoglycans, like chondroitin and dermatan sulfates (hereafter denoted CS/DS) as co-receptors. CS/DS are linear polysaccharides composed of repeating disaccharide units [-4GlcUAb1-3-GalNAc-b1-] and [-4IdoUAa1-3-GalNAc-b1-],which can be sulfated. Uronyl 2-O-sulfotransferase (Ust)introduces sulfation at the C2 of IdoUA and GlcUA resulting inover-sulfated units. Here, we investigated the role of Ust-mediated CS/DS 2-O sulfation in Fgf2-induced cell migration. We found that CHO-K1 cells overexpressing Ust contain significantly more CS/DS2-O sulfated units, whereas Ust knockdown abolished CS/DS 2-O sulfation. These structural differences in CS/DS resulted in altered Fgf2 binding and increased phosphorylation of ERK1/2 (also known as MAPK3 and MAPK1, respectively). As a functional consequence of CS/DS 2-O sulfation and altered Fgf2 binding, cell migration and paxillin activation were increased. Inhibition of sulfation, knockdown of Ust and inhibition of FgfR resulted in reduced migration. Similarly, in 3T3 cells Fgf2 treatment increased migration, which was abolished by Ust knockdown. The proteoglycan controlling the CHO migration was syndecan 1. Knockdown of Sdc1 in CHO-K1 cells overexpressing Ust abolished cell migration.We conclude that the presence of distinctly sulfated CS/DS can tune the Fgf2 effect on cell migration.
Collapse
|
96
|
Wang C, de Jong E, Sjollema KA, Zuhorn IS. Entry of PIP3-containing polyplexes into MDCK epithelial cells by local apical-basal polarity reversal. Sci Rep 2016; 6:21436. [PMID: 26899207 PMCID: PMC4761886 DOI: 10.1038/srep21436] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/22/2016] [Indexed: 02/08/2023] Open
Abstract
The polarized architecture of epithelium presents a barrier to therapeutic drug/gene carriers, which is mainly due to a limited (apical) internalization of the carrier systems. The bacterium Pseudomonas aeruginosa invades epithelial cells by inducing production of apical phosphatidylinositol-3, 4, 5-triphosphate (PIP3), which results in the recruitment of basolateral receptors to the apical membrane. Since basolateral receptors are known receptors for gene delivery vectors, apical PIP3 may improve the internalization of such vectors into epithelial cells. PIP3 and nucleic acids were complexed by the cationic polymer polyethylenimine (PEI), forming PEI/PIP3 polyplexes. PEI/PIP3 polyplexes showed enhanced internalization compared to PEI polyplexes in polarized MDCK cells, while basolateral receptors were found to redistribute and colocalize with PEI/PIP3 polyplexes at the apical membrane. Following their uptake via endocytosis, PEI/PIP3 polyplexes showed efficient endosomal escape. The effectiveness of the PIP3-containing delivery system to generate a physiological effect was demonstrated by an essentially complete knock down of GFP expression in 30% of GFP-expressing MDCK cells following anti-GFP siRNA delivery. Here, we demonstrate that polyplexes can be successfully modified to mimic epithelial entry mechanisms used by Pseudomonas aeruginosa. These findings encourage the development of pathogen-inspired drug delivery systems to improve drug/gene delivery into and across tissue barriers.
Collapse
Affiliation(s)
- Cuifeng Wang
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Edwin de Jong
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Klaas A. Sjollema
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Inge S. Zuhorn
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
97
|
Goker H, Malkan UY, Demiroglu H, Buyukasik Y. Chimeric antigen receptor T cell treatment in hematologic malignancies. Transfus Apher Sci 2016; 54:35-40. [DOI: 10.1016/j.transci.2016.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
98
|
Gopal S, Couchman J, Pocock R. Redefining the role of syndecans in C. elegans biology. WORM 2016; 5:e1142042. [PMID: 27073736 DOI: 10.1080/21624054.2016.1142042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/08/2016] [Indexed: 12/21/2022]
Abstract
Cytosolic calcium is an important factor during fertilization, development and differentiation. Hence, the control of cytosolic calcium levels has been studied extensively for several decades. Numerous calcium channels have been identified and their mechanism of action elucidated. However, the mode of calcium channel regulation remains elusive. Here we discuss our recent findings regarding the role of syndecans in the regulation of cytosolic calcium levels. Syndecans are transmembrane proteoglycans present in both vertebrates and invertebrates that interact with extracellular ligands resulting in the activation of several downstream signaling pathways. We identified a previously unappreciated role of syndecans in cytosolic calcium regulation in mammals that is conserved in C. elegans. We concluded that calcium regulation is the basic, evolutionarily conserved role for syndecans, which enables them to be integral for multiple cellular functions.
Collapse
Affiliation(s)
- Sandeep Gopal
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University , Melbourne, Victoria, Australia
| | - John Couchman
- Department of Biomedical Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University , Melbourne, Victoria, Australia
| |
Collapse
|
99
|
Senadi GC, Liao CM, Kuo KK, Lin JC, Chang LS, Wang JJ, Hu WP. Design, synthesis and antimetastatic evaluation of 1-benzothiazolylphenylbenzotriazoles for photodynamic therapy in oral cancer cells. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00034g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have designed and synthesized a new series of 1-benzothiazolylphenylbenzotriazoles 9a–p and studied their antimetastatic mechanism involved in photosensitive effects induced by UVA in oral cancer cell Ca9-22.
Collapse
|
100
|
Shegefti MS, Malekzadeh M, Malek-Hosseini Z, Khademi B, Ghaderi A, Doroudchi M. Reduced serum levels of syndecan-1 in patients with tongue squamous cell carcinoma. Laryngoscope 2015; 126:E191-5. [PMID: 26667395 DOI: 10.1002/lary.25812] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2015] [Indexed: 11/11/2022]
Abstract
OBJECTIVES/HYPOTHESIS To investigate soluble syndecan-1 (Sdc-1) in the sera of patients with tongue squamous cell carcinoma (SCC) and its correlation with the histopathological criteria of tumors. STUDY DESIGN In a case-control study using a convenient method of sampling, 18 female and 25 male patients with confirmed diagnosis of tongue SCC, and 19 healthy females and 27 males were studied. METHODS Soluble Sdc-1 was measured in the sera of 43 patients with tongue SCC and was compared with that of healthy age-/sex-matched controls using a commercial enzyme-linked immunosorbent assay. Clinical and pathological data, along with the demographic characteristics of the patients, were recorded at the time of sampling. RESULTS The levels of soluble Sdc-1 were decreased in the sera of patients compared to controls (91.17 ± 88.60 vs. 158.17 ± 103.47 ng/mL, P = .002). Although patients who smoke tended to have higher grades (P = .043), there was no significant difference in the level of syndecan-1 between smokers and non-smokers. A significant difference between syndecan-1 in the sera of patients with tumors of different stages was observed (Kruskal-Wallis test P=0.039); however, scarcity of patients in stages I and III decreased the power of the comparison. CONCLUSIONS Sdc-1 levels in the sera of patients do not correlate with the tumor progression in the tongue SCC. This is in contrast with the reported inverse correlation between the expression level of membranous Sdc-1 and the histological grade and size of head and neck tumors. Therefore, Sdc-1 shedding may not be a major mechanism in the progressive loss of its expression regarding tongue tumor. LEVEL OF EVIDENCE NA Laryngoscope, 126:E191-E195, 2016.
Collapse
Affiliation(s)
| | - Mahyar Malekzadeh
- Institute for Cancer Research , School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Malek-Hosseini
- Department of Immunology , School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bijan Khademi
- Department of Otolaryngology , School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Department of Immunology , School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Institute for Cancer Research , School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology , School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Institute for Cancer Research , School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|