1
|
Bugatti A, Zani A, Bardelli M, Giovanetti M, Ravelli C, Ciccozzi M, Caruso A, Caccuri F. Heparan sulfate proteoglycans remodel SARS-CoV-2 spike conformation to allow integrin interaction and infection of endothelial cells. Front Cell Infect Microbiol 2025; 15:1552116. [PMID: 40248367 PMCID: PMC12003327 DOI: 10.3389/fcimb.2025.1552116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
SARS-CoV-2 infects ACE2-negative primary HL-mECs through the interaction of an RGD motif, included in all spike proteins, up to the Omicron BA.1 subvariant, with αvβ3 integrin. Following its entry, SARS-CoV-2 remodels ECs phenotype and promotes angiogenesis in the absence of productive viral replication. Moreover, lack of spike/αvβ3 interaction, occurring in Omicron BA.5 which contains the D405N mutation in the RGD motif, inhibits HL-mECs infection and dysfunction. It is worth noting that anti-spike antibodies do not impact SARS-CoV-2 entry into HL-mECs. This data highlights the fact that i) the RGD motif is not exposed in the entire spike protein and ii) the need of a cofactor favoring spike/αvβ3 interaction. HSPGs are used by different viruses as receptors and coreceptors for their entry into host cells. Here, we use different approaches to scrutinize the role exerted by HSPGs in favoring SARS-CoV-2 infection of ECs. We highlight HSPGs as key molecules responsible for RGD exposure allowing its binding to the αvβ3 integrin as the first step toward viral entry by endocytosis. Indeed, SPR analysis showed lack of spike/αvβ3 interaction in the absence of heparin. This data was further corroborated by immunofluorescence and infectivity assays. Interestingly, the use of Heparinase III or sodium chlorate counteracts the release of proangiogenic molecules and inhibits signaling pathways induced by SARS-CoV-2 infection. Thus, HSPGs may represent a target for preventing SARS-CoV-2 infection of ECs and EC dysfunction-related COVID-19 severity.
Collapse
Affiliation(s)
- Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marta Bardelli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marta Giovanetti
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Cosetta Ravelli
- Section of General Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures, Romania
| |
Collapse
|
2
|
Lee NY, Ture HY, Lee EJ, Jang JA, Kim G, Nam EJ. Syndecan-1 Plays a Role in the Pathogenesis of Sjögren's Disease by Inducing B-Cell Chemotaxis through CXCL13-Heparan Sulfate Interaction. Int J Mol Sci 2024; 25:9375. [PMID: 39273320 PMCID: PMC11394922 DOI: 10.3390/ijms25179375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
In Sjögren's disease (SjD), the salivary glandular epithelial cells can induce the chemotaxis of B cells by secreting B-cell chemokines such as C-X-C motif chemokine ligand 13 (CXCL13). Syndecan-1 (SDC-1) is a major transmembrane heparan sulfate proteoglycan (HSPG) predominantly expressed on epithelial cells that binds to and regulates heparan sulfate (HS)-binding molecules, including chemokines. We aimed to determine whether SDC-1 plays a role in the pathogenesis of SjD by acting on the binding of HS to B-cell chemokines. To assess changes in glandular inflammation and SDC-1 concentrations in the submandibular gland (SMG) and blood, female NOD/ShiLtJ and sex- and age-matched C57BL/10 mice were used. In the SMG of NOD/ShiLtJ mice, inflammatory responses were identified at 8 weeks of age, but increased SDC-1 concentrations in the SMG and blood were observed at 6 weeks of age, when inflammation had not yet started. As the inflammation of the SMG worsened, the SDC-1 concentrations in the SMG and blood increased. The expression of the CXCL13 and its receptor C-X-C chemokine receptor type 5 (CXCR5) began to increase in the SMG at 6 weeks of age and continued until 12 weeks of age. Immunofluorescence staining in SMG tissue and normal murine mammary gland cells confirmed the co-localization of SDC-1 and CXCL13, and SDC-1 formed a complex with CXCL13 in an immunoprecipitation assay. Furthermore, NOD/ShiLtJ mice were treated with 5 mg/kg HS intraperitoneally thrice per week for 6-10 weeks of age, and the therapeutic effects in the SMG were assessed at the end of 10 weeks of age. NOD/ShiLtJ mice treated with HS showed attenuated salivary gland inflammation with reduced B-cell infiltration, germinal center formation and CXCR5 expression. These findings suggest that SDC-1 plays a pivotal role in the pathogenesis of SjD by binding to CXCL13 through the HS chain.
Collapse
Affiliation(s)
- Nan Young Lee
- Department of Clinical Pathology, School of Medicine, Kyungpook National University, Daegu 41405, Republic of Korea
| | - Hirut Yadeta Ture
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41405, Republic of Korea
| | - Eun Ju Lee
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu Fatima Hospital, Daegu 41199, Republic of Korea
| | - Ji Ae Jang
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu Fatima Hospital, Daegu 41199, Republic of Korea
| | - Gunwoo Kim
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu Fatima Hospital, Daegu 41199, Republic of Korea
| | - Eon Jeong Nam
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41405, Republic of Korea
| |
Collapse
|
3
|
Vazaios K, Stavrakaki Ε, Vogelezang LB, Ju J, Waranecki P, Metselaar DS, Meel MH, Kemp V, van den Hoogen BG, Hoeben RC, Chiocca EA, Goins WF, Stubbs A, Li Y, Alonso MM, Calkoen FG, Hulleman E, van der Lugt J, Lamfers ML. The heterogeneous sensitivity of pediatric brain tumors to different oncolytic viruses is predicted by unique gene expression profiles. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200804. [PMID: 38694569 PMCID: PMC11060958 DOI: 10.1016/j.omton.2024.200804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/11/2024] [Indexed: 05/04/2024]
Abstract
Despite decades of research, the prognosis of high-grade pediatric brain tumors (PBTs) remains dismal; however, recent cases of favorable clinical responses were documented in clinical trials using oncolytic viruses (OVs). In the current study, we employed four different species of OVs: adenovirus Delta24-RGD, herpes simplex virus rQNestin34.5v1, reovirus R124, and the non-virulent Newcastle disease virus rNDV-F0-GFP against three entities of PBTs (high-grade gliomas, atypical teratoid/rhabdoid tumors, and ependymomas) to determine their in vitro efficacy. These four OVs were screened on 14 patient-derived PBT cell cultures and the degree of oncolysis was assessed using an ATP-based assay. Subsequently, the observed viral efficacies were correlated to whole transcriptome data and Gene Ontology analysis was performed. Although no significant tumor type-specific OV efficacy was observed, the analysis revealed the intrinsic biological processes that associated with OV efficacy. The predictive power of the identified expression profiles was further validated in vitro by screening additional PBTs. In summary, our results demonstrate OV susceptibility of multiple patient-derived PBT entities and the ability to predict in vitro responses to OVs using unique expression profiles. Such profiles may hold promise for future OV preselection with effective oncolytic potency in a specific tumor, therewith potentially improving OV responses.
Collapse
Affiliation(s)
- Konstantinos Vazaios
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
- Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Εftychia Stavrakaki
- Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Lisette B. Vogelezang
- Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Jie Ju
- Department of Pathology and Clinical Bioinformatics, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Piotr Waranecki
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Dennis S. Metselaar
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Michaël H. Meel
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
- Department of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Vera Kemp
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | | | - Rob C. Hoeben
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - E. Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - William F. Goins
- Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Dr, Pittsburgh, PA 15219, USA
| | - Andrew Stubbs
- Department of Pathology and Clinical Bioinformatics, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Marta M. Alonso
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), Avda. de Pío XII, 55, 31008 Pamplona, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Av. de Pío XII, 36, 31008 Pamplona, Spain
- Health Research Institute of Navarra (IDISNA), Av. de Pío XII, 36, 31008 Pamplona, Spain
| | - Friso G. Calkoen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Jasper van der Lugt
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Martine L.M. Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| |
Collapse
|
4
|
Gagan S, Khapuinamai A, Kapoor D, Sharma P, Yadavalli T, Joseph J, Shukla D, Bagga B. Exploring Heparanase Levels in Tears: Insights From Herpes Simplex Virus-1 Keratitis Patients and Animal Studies. Invest Ophthalmol Vis Sci 2024; 65:7. [PMID: 38466284 DOI: 10.1167/iovs.65.3.7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Purpose Heparanase (HPSE) cleaves heparan sulfate proteoglycans during herpes simplex virus-1 (HSV-1) infection, aiding in viral egress and disease progression. Its action has been well established in in vitro and in vivo models, but its relevance in human patients remains unclear. This study aimed to specifically evaluate tear HPSE levels of patients with herpes simplex keratitis (HSK) and to correlate these findings with a commonly used murine model. Methods Tear samples from patient and mice samples were collected at LV Prasad Eye Institute, Hyderabad, India, and at the University of Illinois, Chicago, IL, respectively. Tears were collected from HSV-1 patients, bacterial/fungal keratitis cases, and healthy individuals. For in vivo study, C57BL/6 mice were infected with HSV-1 (McKrae strain) followed by tear fluid collection at various time points (0-10 days). Results The HSV-1, bacterial keratitis, fungal keratitis, and healthy control groups each had 30 patients. There was a significant difference in HPSE expression in the HSV-1 infected eyes (1.55 ± 0.19 units/mL) compared to HSV-1 contralateral eyes (1.23 ± 0.13 units/mL; P = 0.82), bacterial keratitis eyes (0.87 ± 0.15 units/mL; P = 0.0078), fungal keratitis eyes (0.64 ± 0.09 units/mL; P < 0.00001), and normal controls (0.53 ± 0.06 units/mL; P < 0.00001). C57BL/6 mice tear HPSE expression in infected eyes was 0.66 to 5.57 ng heparan sulfate (HS) removed per minute when compared to non-infected eye (range, 0.70-3.67 ng HS removed per minute). Conclusions To the best of our knowledge, this study is the first to report elevated HPSE levels in the tears of patients with different forms of HSV-1 keratitis, and it confirms similar findings in a murine model, providing a valuable basis for future in vivo and clinical research on HSV-1 ocular infection.
Collapse
Affiliation(s)
- Satyashree Gagan
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
- Manipal Academy of Higher Education, Karnataka, India
| | - Agimanailiu Khapuinamai
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Divya Kapoor
- Department of Ophthalmology and Visual Science, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Pankaj Sharma
- Department of Ophthalmology and Visual Science, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Science, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Deepak Shukla
- Department of Ophthalmology and Visual Science, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Bhupesh Bagga
- Shantilal Shanghvi Cornea Institute, The Ramoji Foundation Centre for Ocular Infections, LV Prasad Eye Institute, Hyderabad, Telangana, India
| |
Collapse
|
5
|
Rajan R, Hanifah M, Mariappan V, Anand M, Balakrishna Pillai A. Soluble Endoglin and Syndecan-1 levels predicts the clinical outcome in COVID-19 patients. Microb Pathog 2024; 188:106558. [PMID: 38272329 DOI: 10.1016/j.micpath.2024.106558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Endothelial instability is reported to be involved in the pathogenesis of COVID-19. The mechanism that regulates the endothelial dysfunction and disease virulence is not known. Studies on proteins that are released into circulation by activated endothelial cells may provide some means to understand the disease manifestation. The study investigated the circulating levels of two molecules Endoglin (Eng) and Syndecan-1 (SDC-1) that are presumed to be involved in the maintenance of endothelial integrity and their association with hypercoagulation marker in COVID-19 patients. The serum levels of Eng, SDC-1, D-mer were evaluated using ELISA at the time of admission (DOA) and day 7 post-admission among COVID-19 patients (N = 39 with 17 moderate and 22 severe cases). Compared to the time of admission, there was an increase in sEng and sSDC1 levels in all COVID-19 cases on day 7 post admission. The serum levels of sEng and sSDC-1 was significantly (P ≤ 0.001 & P ≤ 0.01 respectively) elevated in severe cases including the four deceased group compared to moderate cases on day 7 post admission. Further, the study molecules showed a strong positive association (P ≤ 0.001) with the hypercoagulation marker D-mer. The results show an early shedding of the endothelial proteins sEng and sSDC-1 into circulation as a host response to the viral infection during the febrile phase of infection. Increased levels of sEng and sSDC-1 along with D-mer could be beneficial in predicting COVID-19 disease severity.
Collapse
Affiliation(s)
- Remya Rajan
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Mohamed Hanifah
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Monica Anand
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| |
Collapse
|
6
|
Zhou W, Hong J, Han J, Cai F, Tang Q, Yu Q, Li G, Ma S, Liu X, Huo S, Chen K, Zhu F. Silkworm glycosaminoglycans bind to Bombyx mori nuclear polyhedrosis virus and facilitate its entry. Int J Biol Macromol 2023; 253:127352. [PMID: 37838120 DOI: 10.1016/j.ijbiomac.2023.127352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/29/2023] [Accepted: 10/04/2023] [Indexed: 10/16/2023]
Abstract
Interacting with cell surface attachment factors or receptors is the first step for virus infection. Glycans cover a thick layer on eukaryotic cells and are potential targets of various viruses. Bombyx mori nuclear polyhedrosis viruses (BmNPV) is a baculovirus that causes huge economic loss to the sericulture industry but the mechanism of infection is unclear. Looking for potential host receptors for the virus is an important task. In this study, we investigated the role of glycosaminoglycan (GAG) modifications, including heparan sulfate (HS) and chondroitin sulfate (CS), during BmNPV infection. Enzymatic removal of cell surface HS and CS effectively inhibited BmNPV infection and replication. Exogenous HS and CS can directly bind to BmNPV virion in solution and act as neutralizers for viral infection. Furthermore, the expression of enzymes involved in GAG biosynthesis was upregulated in the BmNPV susceptible silkworm after virus administration, but down-regulated in the resistant strain after virus treatment, suggesting that BmNPV was able to utilize host cell machinery to promote the biosynthesis of GAGs. This study demonstrated HS and CS as important attachment factors that facilitate the viral entry process, and targeting HS and CS can be an effective means of inhibiting BmNPV infection.
Collapse
Affiliation(s)
- Weiwei Zhou
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Jindie Hong
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Jinying Han
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Fuchuan Cai
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Qi Tang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Qian Yu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Guohui Li
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Shangshang Ma
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoyong Liu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Shuhao Huo
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Feifei Zhu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
7
|
Ghondaghsaz E, Khalaji A, Norouzi M, Fraser DD, Alilou S, Behnoush AH. The utility of syndecan-1 circulating levels as a biomarker in patients with previous or active COVID-19: a systematic review and meta-analysis. BMC Infect Dis 2023; 23:510. [PMID: 37542221 PMCID: PMC10401738 DOI: 10.1186/s12879-023-08473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND With the emergence of coronavirus disease of 2019 (COVID-19), several blood biomarkers have been identified, including the endothelial biomarker syndecan-1, a surface proteoglycan. In the current systematic review and meta-analysis, we aimed to assess the diagnostic and prognostic role of syndecan-1 in COVID-19. METHODS PubMed, Embase, Scopus, and Web of Science, as international databases, were searched for relevant studies measuring blood syndecan-1 levels in COVID-19 patients, COVID-19 convalescents, and healthy control subjects, in patients with different COVID-19 severities and/or in COVID-19 patients with poor outcomes. Random-effect meta-analysis was performed using STATA to calculate the standardized mean difference (SMD) and 95% confidence interval (CI) for the comparison between COVID-19 patients and healthy control subjects or COVID-19 convalescents and controls. RESULTS After screening by title/abstract and full text, 17 studies were included in the final review. Meta-analysis of syndecan-1 levels in COVID-19 compared with healthy control subjects revealed that patients with COVID-19 had significantly higher syndecan-1 levels (SMD 1.53, 95% CI 0.66 to 2.41, P < 0.01). In contrast, COVID-19 convalescent patients did not show significant difference with non-convalescents (SMD 0.08, 95% CI -0.63 to 0.78, P = 0.83). Regarding disease severity, two studies reported that more severe forms of the disease were associated with increased syndecan-1 levels. Moreover, patients who died from COVID-19 had higher syndecan-1 levels compared with survivors (SMD 1.22, 95% CI 0.10 to 2.33, P = 0.03). CONCLUSION Circulating syndecan-1 level can be used as a biomarker of endothelial dysfunction in COVID-19, as it was increased in COVID-19 patients and was higher in more severe instances of the disease. Further larger studies are needed to confirm these findings and further enlighten the role of syndecan-1 in clinical settings.
Collapse
Affiliation(s)
- Elina Ghondaghsaz
- Undergraduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Amirmohammad Khalaji
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151, Iran
| | - Mitra Norouzi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Douglas D Fraser
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Department of Pediatrics, Western University, London, ON, Canada
- Department of Physiology & Pharmacology, Western University, London, ON, Canada
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada
| | - Sanam Alilou
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Behnoush
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151, Iran.
| |
Collapse
|
8
|
Jasim SA, Mahdi RS, Bokov DO, Najm MAA, Sobirova GN, Bafoyeva ZO, Taifi A, Alkadir OKA, Mustafa YF, Mirzaei R, Karampoor S. The deciphering of the immune cells and marker signature in COVID-19 pathogenesis: An update. J Med Virol 2022; 94:5128-5148. [PMID: 35835586 PMCID: PMC9350195 DOI: 10.1002/jmv.28000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/28/2022] [Accepted: 07/13/2022] [Indexed: 12/15/2022]
Abstract
The precise interaction between the immune system and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical in deciphering the pathogenesis of coronavirus disease 2019 (COVID-19) and is also vital for developing novel therapeutic tools, including monoclonal antibodies, antivirals drugs, and vaccines. Viral infections need innate and adaptive immune reactions since the various immune components, such as neutrophils, macrophages, CD4+ T, CD8+ T, and B lymphocytes, play different roles in various infections. Consequently, the characterization of innate and adaptive immune reactions toward SARS-CoV-2 is crucial for defining the pathogenicity of COVID-19. In this study, we explain what is currently understood concerning the conventional immune reactions to SARS-CoV-2 infection to shed light on the protective and pathogenic role of immune response in this case. Also, in particular, we investigate the in-depth roles of other immune mediators, including neutrophil elastase, serum amyloid A, and syndecan, in the immunopathogenesis of COVID-19.
Collapse
Affiliation(s)
| | - Roaa Salih Mahdi
- Department of Pathology, College of MedicineUniversity of BabylonHillaIraq
| | - Dmitry Olegovich Bokov
- Institute of PharmacySechenov First Moscow State Medical UniversityMoscowRussian Federation
- Laboratory of Food ChemistryFederal Research Center of Nutrition, Biotechnology and Food SafetyMoscowRussian Federation
| | - Mazin A. A. Najm
- Pharmaceutical Chemistry Department, College of PharmacyAl‐Ayen UniversityThi‐QarIraq
| | - Guzal N. Sobirova
- Department of Rehabilitation, Folk Medicine and Physical EducationTashkent Medical AcademyTashkentUzbekistan
| | - Zarnigor O. Bafoyeva
- Department of Rehabilitation, Folk Medicine and Physical EducationTashkent Medical AcademyTashkentUzbekistan
| | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of PharmacyUniversity of MosulMosulIraq
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research CenterIran University of Medical SciencesTehranIran
| |
Collapse
|
9
|
Sharma P, Kapoor D, Shukla D. Role of Heparanase and Syndecan-1 in HSV-1 Release from Infected Cells. Viruses 2022; 14:2156. [PMID: 36298711 PMCID: PMC9612286 DOI: 10.3390/v14102156] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022] Open
Abstract
Herpes Simplex Virus 1 (HSV-1) is a neurotropic human virus that belongs to the Alphaherpesvirinae subfamily of Herpesviridae. Establishment of its productive infection and progression of disease pathologies depend largely on successful release of virions from the virus-producing cells. HSV-1 is known to exploit many host factors for its release. Recent studies have shown that heparanase (HPSE) is one such host enzyme that is recruited for this purpose. It is an endoglycosidase that cleaves heparan sulfate (HS) from the surface of infected cells. HS is a virus attachment coreceptor that is commonly found on cell surfaces as HS proteoglycans e.g., syndecan-1 (SDC-1). The current model suggests that HSV-1 during the late stage of infection upregulates HPSE, which in turn enhances viral release by removing the virus-trapping HS moieties. In addition to its role in directly enabling viral release, HPSE accelerates the shedding of HS-containing ectodomains of SDC-1, which enhances HSV-1 release via a similar mechanism by upregulating CREB3 and COPII proteins. This review outlines the role of HPSE and SDC-1 as newly assigned host factors that facilitate HSV-1 release during a lytic infection cycle.
Collapse
Affiliation(s)
- Pankaj Sharma
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Divya Kapoor
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
10
|
Ramos-Martínez IE, Ramos-Martínez E, Segura-Velázquez RÁ, Saavedra-Montañez M, Cervantes-Torres JB, Cerbón M, Papy-Garcia D, Zenteno E, Sánchez-Betancourt JI. Heparan Sulfate and Sialic Acid in Viral Attachment: Two Sides of the Same Coin? Int J Mol Sci 2022; 23:ijms23179842. [PMID: 36077240 PMCID: PMC9456526 DOI: 10.3390/ijms23179842] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 12/11/2022] Open
Abstract
Sialic acids and heparan sulfates make up the outermost part of the cell membrane and the extracellular matrix. Both structures are characterized by being negatively charged, serving as receptors for various pathogens, and are highly expressed in the respiratory and digestive tracts. Numerous viruses use heparan sulfates as receptors to infect cells; in this group are HSV, HPV, and SARS-CoV-2. Other viruses require the cell to express sialic acids, as is the case in influenza A viruses and adenoviruses. This review aims to present, in a general way, the participation of glycoconjugates in viral entry, and therapeutic strategies focused on inhibiting the interaction between the virus and the glycoconjugates. Interestingly, there are few studies that suggest the participation of both glycoconjugates in the viruses addressed here. Considering the biological redundancy that exists between heparan sulfates and sialic acids, we propose that it is important to jointly evaluate and design strategies that contemplate inhibiting the interactions of both glycoconjugates. This approach will allow identifying new receptors and lead to a deeper understanding of interspecies transmission.
Collapse
Affiliation(s)
- Ivan Emmanuel Ramos-Martínez
- Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Edgar Ramos-Martínez
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - René Álvaro Segura-Velázquez
- Unidad de Investigación, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Manuel Saavedra-Montañez
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Jacquelynne Brenda Cervantes-Torres
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Dulce Papy-Garcia
- Glycobiology, Cell Growth ant Tissue Repair Research Unit (Gly-CRRET), Université Paris Est Créteil (UPEC), F-94010 Créteil, France
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - José Ivan Sánchez-Betancourt
- Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Correspondence:
| |
Collapse
|
11
|
Abidine Y, Liu L, Wallén O, Trybala E, Olofsson S, Bergström T, Bally M. Cellular Chondroitin Sulfate and the Mucin-like Domain of Viral Glycoprotein C Promote Diffusion of Herpes Simplex Virus 1 While Heparan Sulfate Restricts Mobility. Viruses 2022; 14:v14081836. [PMID: 36016458 PMCID: PMC9412521 DOI: 10.3390/v14081836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 01/26/2023] Open
Abstract
The diffusion of viruses at the cell membrane is essential to reach a suitable entry site and initiate subsequent internalization. Although many viruses take advantage of glycosaminoglycans (GAG) to bind to the cell surface, little is known about the dynamics of the virus–GAG interactions. Here, single-particle tracking of the initial interaction of individual herpes simplex virus 1 (HSV-1) virions reveals a heterogeneous diffusive behavior, regulated by cell-surface GAGs with two main diffusion types: confined and normal free. This study reports that different GAGs can have competing influences in mediating diffusion on the cells used here: chondroitin sulfate (CS) enhances free diffusion but hinders virus attachment to cell surfaces, while heparan sulfate (HS) promotes virus confinement and increases entry efficiency. In addition, the role that the viral mucin-like domains (MLD) of the HSV-1 glycoprotein C plays in facilitating the diffusion of the virus and accelerating virus penetration into cells is demonstrated. Together, our results shed new light on the mechanisms of GAG-regulated virus diffusion at the cell surface for optimal internalization. These findings may be extendable to other GAG-binding viruses.
Collapse
Affiliation(s)
- Yara Abidine
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, SE-90185 Umeå, Sweden
| | - Lifeng Liu
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, SE-90185 Umeå, Sweden
| | - Oskar Wallén
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
| | - Edward Trybala
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, SE-41346 Göteborg, Sweden
| | - Sigvard Olofsson
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, SE-41346 Göteborg, Sweden
| | - Tomas Bergström
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, SE-41346 Göteborg, Sweden
| | - Marta Bally
- Department of Clinical Microbiology, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, SE-90185 Umeå, Sweden
- Correspondence:
| |
Collapse
|
12
|
Chang K, Majmudar H, Tandon R, Volin MV, Tiwari V. Induction of Filopodia During Cytomegalovirus Entry Into Human Iris Stromal Cells. Front Microbiol 2022; 13:834927. [PMID: 35450284 PMCID: PMC9018114 DOI: 10.3389/fmicb.2022.834927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/07/2022] [Indexed: 12/25/2022] Open
Abstract
Many viruses exploit thin projections of filopodia for cell entry and cell-to-cell spread. Using primary cultures of human iris stromal (HIS) cells derived from human eye donors, we report a significant increase in filopodia formation during human cytomegalovirus (HCMV) infection. Using confocal microscopy, we observed a large number of virions being frequently associated along the filopodia prior to cell infection. Depolymerization of actin filaments resulted in a significant inhibition of HCMV entry into HIS cell. Our results further revealed that the transient expression of HCMV envelope glycoprotein B (gB) triggers the induction of the filopodial system. Since gB is known to bind the diverse chains of heparan sulfate (HS), a comparative study was performed to evaluate the gB-mediated filopodial induction in cells expressing either wild-type HS and/or 3-O sulfated HS (3-OS HS). We found that cells co-expressing HCMV gB together with the 3-O sulfotranseferase-3 (3-OST-3) enzyme had a much higher and robust filopodia induction compared to cells co-expressing gB with wild-type HS. The above results were further verified by pre-treating HIS cells with anti-3-OS HS (G2) peptide and/or heparinase-I before challenging with HCMV infection, which resulted in a significant loss in the filopodial counts as well as decreased viral infectivity. Taken together, our findings highlight that HCMV entry into HIS cells actively modulates the actin cytoskeleton via coordinated actions possibly between gB and the 3-OS HS receptor to influence viral infectivity.
Collapse
Affiliation(s)
- Kenneth Chang
- Department of Microbiology and Immunology, College of Graduate Studies, Chicago College of Osteopathic Medicine, and Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Hardik Majmudar
- Department of Microbiology and Immunology, College of Graduate Studies, Chicago College of Osteopathic Medicine, and Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Ritesh Tandon
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Michael V Volin
- Department of Microbiology and Immunology, College of Graduate Studies, Chicago College of Osteopathic Medicine, and Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, College of Graduate Studies, Chicago College of Osteopathic Medicine, and Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| |
Collapse
|
13
|
Quinn CH, Beierle AM, Hutchins SC, Marayati R, Bownes LV, Stewart JE, Markert HR, Erwin MH, Aye JM, Yoon KJ, Friedman GK, Willey CD, Markert JM, Beierle EA. Targeting High-Risk Neuroblastoma Patient-Derived Xenografts with Oncolytic Virotherapy. Cancers (Basel) 2022; 14:cancers14030762. [PMID: 35159029 PMCID: PMC8834037 DOI: 10.3390/cancers14030762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is the leading cause of death by disease in children, and over 15% of pediatric cancer-related mortalities are due to neuroblastoma. Current treatment options for neuroblastoma remain suboptimal as they often have significant toxicities, are associated with long-term side effects, and result in disease relapse in over half of children with high-risk disease. There is a dire need for new therapies, and oncolytic viruses may represent an effective solution. Oncolytic viruses attack tumor cells in two ways: direct infection of tumor cells leading to cytolysis, and production of a debris field that stimulates an anti-tumor immune response. Our group has previously shown that M002, an oncolytic herpes simplex virus (oHSV), genetically engineered to express murine interleukin-12 (mIL-12), was effective at targeting and killing long term passage tumor cell lines. In the current study, we investigated M002 in three neuroblastoma patient-derived xenografts (PDXs). PDXs better recapitulate the human condition, and these studies were designed to gather robust data for translation to a clinical trial. We found that all three PDXs expressed viral entry receptors, and that the virus actively replicated in the cells. M002 caused significant tumor cell death in 2D culture and 3D bioprinted tumor models. Finally, the PDXs displayed variable susceptibility to M002, with a more profound effect on high-risk neuroblastoma PDXs compared to low-risk PDX. These findings validate the importance of incorporating PDXs for preclinical testing of oncolytic viral therapeutics and showcase a novel technique, 3D bioprinting, to test therapies in PDXs. Collectively, our data indicate that oHSVs effectively target high-risk neuroblastoma, and support the advancement of this therapy to the clinical setting.
Collapse
Affiliation(s)
- Colin H. Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (C.H.Q.); (R.M.); (L.V.B.); (J.E.S.); (H.R.M.); (M.H.E.)
| | - Andee M. Beierle
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.M.B.); (C.D.W.)
| | - Sara Claire Hutchins
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.C.H.); (J.M.A.); (G.K.F.)
| | - Raoud Marayati
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (C.H.Q.); (R.M.); (L.V.B.); (J.E.S.); (H.R.M.); (M.H.E.)
| | - Laura V. Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (C.H.Q.); (R.M.); (L.V.B.); (J.E.S.); (H.R.M.); (M.H.E.)
| | - Jerry E. Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (C.H.Q.); (R.M.); (L.V.B.); (J.E.S.); (H.R.M.); (M.H.E.)
| | - Hooper R. Markert
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (C.H.Q.); (R.M.); (L.V.B.); (J.E.S.); (H.R.M.); (M.H.E.)
| | - Michael H. Erwin
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (C.H.Q.); (R.M.); (L.V.B.); (J.E.S.); (H.R.M.); (M.H.E.)
| | - Jamie M. Aye
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.C.H.); (J.M.A.); (G.K.F.)
| | - Karina J. Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Gregory K. Friedman
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (S.C.H.); (J.M.A.); (G.K.F.)
| | - Christopher D. Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.M.B.); (C.D.W.)
| | - James M. Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Elizabeth A. Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35205, USA; (C.H.Q.); (R.M.); (L.V.B.); (J.E.S.); (H.R.M.); (M.H.E.)
- Correspondence: ; Tel.: +1-205-638-9688
| |
Collapse
|
14
|
Shukla SD, Valyi-Nagy T. Host Molecules That Promote Pathophysiology of Ocular Herpes. Front Microbiol 2022; 13:818658. [PMID: 35145504 PMCID: PMC8822155 DOI: 10.3389/fmicb.2022.818658] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/03/2022] [Indexed: 11/23/2022] Open
Abstract
Herpes simplex virus type-1 (HSV-1) is a human virus that causes lifelong infections in a large population worldwide. Recurrence of HSV-1 from latency in trigeminal ganglion (TG) is the trigger of the morbidities seen with this virus. In addition to causing fever blisters and cold sores, occasionally the virus can also cause corneal lesions resulting in blindness in untreated individuals. Several host cell proteins play important roles in HSV-1 infection of the eye. HSV-1 enters into the corneal epithelial cells via its interactions with cell surface receptors. In parallel, the Toll-like receptors sense viral invasion and activate defense mechanisms to fight the infection. New data shows that Optineurin, a host autophagy receptor is also activated to degrade viral particles. In contrast, activation of heparanase, a host enzyme, induces an immune-inflammatory response, which triggers pro-inflammatory and pro-angiogenic environment and ultimately results in many of the clinical features seen with HSV-1 infection of the cornea. Rarely, HSV-1 can also spread to the central nervous system causing serious diseases. In this review, we summarize the latest knowledge on host molecules that promote pathophysiological aspects of ocular herpes.
Collapse
Affiliation(s)
- Sajal Deea Shukla
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Illinois Mathematics and Science Academy, Aurora, IL, United States
| | - Tibor Valyi-Nagy
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Tibor Valyi-Nagy,
| |
Collapse
|
15
|
Bermejo‐Jambrina M, Eder J, Kaptein TM, van Hamme JL, Helgers LC, Vlaming KE, Brouwer PJM, van Nuenen AC, Spaargaren M, de Bree GJ, Nijmeijer BM, Kootstra NA, van Gils MJ, Sanders RW, Geijtenbeek TBH. Infection and transmission of SARS-CoV-2 depend on heparan sulfate proteoglycans. EMBO J 2021; 40:e106765. [PMID: 34510494 PMCID: PMC8521309 DOI: 10.15252/embj.2020106765] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 12/27/2022] Open
Abstract
The current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and outbreaks of new variants highlight the need for preventive treatments. Here, we identified heparan sulfate proteoglycans as attachment receptors for SARS-CoV-2. Notably, neutralizing antibodies against SARS-CoV-2 isolated from COVID-19 patients interfered with SARS-CoV-2 binding to heparan sulfate proteoglycans, which might be an additional mechanism of antibodies to neutralize infection. SARS-CoV-2 binding to and infection of epithelial cells was blocked by low molecular weight heparins (LMWH). Although dendritic cells (DCs) and mucosal Langerhans cells (LCs) were not infected by SARS-CoV-2, both DC subsets efficiently captured SARS-CoV-2 via heparan sulfate proteoglycans and transmitted the virus to ACE2-positive cells. Notably, human primary nasal cells were infected by SARS-CoV-2, and infection was blocked by pre-treatment with LMWH. These data strongly suggest that heparan sulfate proteoglycans are important attachment receptors facilitating infection and transmission, and support the use of LMWH as prophylaxis against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Marta Bermejo‐Jambrina
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Julia Eder
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Tanja M Kaptein
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - John L van Hamme
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Leanne C Helgers
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Killian E Vlaming
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Philip J M Brouwer
- Department of Medical MicrobiologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ad C van Nuenen
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marcel Spaargaren
- Department of Pathology, Lymphoma and Myeloma Center Amsterdam (LYMMCARE)Cancer Center Amsterdam (CCA)Amsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Godelieve J de Bree
- Department of Internal MedicineAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Bernadien M Nijmeijer
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Neeltje A Kootstra
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marit J van Gils
- Department of Medical MicrobiologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Rogier W Sanders
- Department of Medical MicrobiologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Microbiology and ImmunologyWeill Medical College of Cornell UniversityNew YorkNYUSA
| | - Teunis B H Geijtenbeek
- Department of Experimental ImmunologyAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
16
|
Karasneh GA, Kapoor D, Bellamkonda N, Patil CD, Shukla D. Protease, Growth Factor, and Heparanase-Mediated Syndecan-1 Shedding Leads to Enhanced HSV-1 Egress. Viruses 2021; 13:v13091748. [PMID: 34578329 DOI: 10.3390/v13091748] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 12/29/2022] Open
Abstract
Heparan sulfate (HS) and heparan sulfate proteoglycans (HSPGs) are considered important for the entry of many different viruses. Previously, we demonstrated that heparanase (HPSE), the host enzyme responsible for cleaving HS chains, is upregulated by herpes simplex virus-1 (HSV-1) infection. Higher levels of HPSE accelerate HS removal from the cell surface, facilitating viral release from infected cells. Here, we study the effects of overexpressing HPSE on viral entry, cell-to-cell fusion, plaque formation, and viral egress. We provide new information that higher levels of HPSE reduce syncytial plaque formation while promoting egress and extracellular release of the virions. We also found that transiently enhanced expression of HPSE did not affect HSV-1 entry into host cells or HSV-1-induced cell-to-cell fusion, suggesting that HPSE activation is tightly regulated and facilitates extracellular release of the maturing virions. We demonstrate that an HSPG-shedding agonist, PMA; a protease, thrombin; and a growth factor, EGF as well as bacterially produced recombinant heparinases resulted in enhanced HSV-1 release from HeLa and human corneal epithelial (HCE) cells. Our findings here underscore the significance of syndecan-1 functions in the HSV-1 lifecycle, provide evidence that the shedding of syndecan-1 ectodomain is another way HPSE works to facilitate HSV-1 release, and add new evidence on the significance of various HSPG shedding agonists in HSV-1 release from infected cells.
Collapse
Affiliation(s)
- Ghadah A Karasneh
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1855 W. Taylor, Chicago, IL 60612, USA
| | - Divya Kapoor
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1855 W. Taylor, Chicago, IL 60612, USA
| | - Navya Bellamkonda
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1855 W. Taylor, Chicago, IL 60612, USA
| | - Chandrashekhar D Patil
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1855 W. Taylor, Chicago, IL 60612, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1855 W. Taylor, Chicago, IL 60612, USA
| |
Collapse
|
17
|
Karampoor S, Zahednasab H, Farahmand M, Mirzaei R, Zamani F, Tabibzadeh A, Bouzari B, Ajdarkosh H, Nikkhah M, Hashemi MR, Laali A, Keyvani H. A possible pathogenic role of Syndecan-1 in the pathogenesis of coronavirus disease 2019 (COVID-19). Int Immunopharmacol 2021; 97:107684. [PMID: 33932696 PMCID: PMC8052477 DOI: 10.1016/j.intimp.2021.107684] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023]
Abstract
A cell-surface heparan proteoglycan called Syndecan-1 (SDC-1) has multiple roles in healthy and pathogenic conditions, including respiratory viral infection. In this study, we explore the dynamic alternation in the levels of SDC-1 in cases with COVID-19. A total of 120 cases definitely diagnosed with COVID-19 were admitted to the Firoozgar Hospital, Tehran, Iran, from December 1, 2020, to January 29, 2021, and included in our study. Also, 58 healthy subjects (HS) were chosen as the control group. Patients were classified into two groups: 1) ICU patients and (63 cases) 2) non-ICU patients (57 cases). The dynamic changes of serum SCD-1, CRP, IL-6, IL-10, IL-18, and Vit D levels a well as the disease activity were investigated in three-time points (T1-T3). Our results indicated that the COVID-19 patients had significantly increased SCD-1, CRP, IL-6, IL-10, and IL-18 levels than in HS, while the Vit D levels in COVID-19 patients were significantly lower than HS. Further analysis demonstrated that the SCD-1, CRP, IL-6, IL-10, and IL-18 levels in ICU patients were significantly higher than in non-ICU patients. Tracking dynamic changes in the above markers indicated that on the day of admission, the SCD-1, CRP, IL-6, IL-10, and IL-18 levels were gradually increased on day 5 (T2) and then gradually decreased on day 10 (T3). ROC curve analysis suggests that markers mentioned above, SDC-1, IL-6, and IL-18 are valuable indicators in evaluating the activity of COVID-19. All in all, it seems that the serum SDC-1 levels alone or combined with other markers might be a good candidate for disease activity monitoring.
Collapse
Affiliation(s)
- Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hamid Zahednasab
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mohammad Farahmand
- Department of Medical Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Tabibzadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Bouzari
- Department of Pathology, Firouzgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Ajdarkosh
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Nikkhah
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Melika Razavi Hashemi
- Department of Pathology, Firouzgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Laali
- Department of Infectious Disease, School of Medicine, Firoozgar General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Keyvani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Heparan Sulfate Proteoglycans in Viral Infection and Treatment: A Special Focus on SARS-CoV-2. Int J Mol Sci 2021; 22:ijms22126574. [PMID: 34207476 PMCID: PMC8235362 DOI: 10.3390/ijms22126574] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/27/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) encompass a group of glycoproteins composed of unbranched negatively charged heparan sulfate (HS) chains covalently attached to a core protein. The complex HSPG biosynthetic machinery generates an extraordinary structural variety of HS chains that enable them to bind a plethora of ligands, including growth factors, morphogens, cytokines, chemokines, enzymes, matrix proteins, and bacterial and viral pathogens. These interactions translate into key regulatory activity of HSPGs on a wide range of cellular processes such as receptor activation and signaling, cytoskeleton assembly, extracellular matrix remodeling, endocytosis, cell-cell crosstalk, and others. Due to their ubiquitous expression within tissues and their large functional repertoire, HSPGs are involved in many physiopathological processes; thus, they have emerged as valuable targets for the therapy of many human diseases. Among their functions, HSPGs assist many viruses in invading host cells at various steps of their life cycle. Viruses utilize HSPGs for the attachment to the host cell, internalization, intracellular trafficking, egress, and spread. Recently, HSPG involvement in the pathogenesis of SARS-CoV-2 infection has been established. Here, we summarize the current knowledge on the molecular mechanisms underlying HSPG/SARS-CoV-2 interaction and downstream effects, and we provide an overview of the HSPG-based therapeutic strategies that could be used to combat such a fearsome virus.
Collapse
|
19
|
Mariappan V, Adikari S, Shanmugam L, Easow JM, Balakrishna Pillai A. Expression dynamics of vascular endothelial markers: endoglin and syndecan-1 in predicting dengue disease outcome. Transl Res 2021; 232:121-141. [PMID: 33567345 DOI: 10.1016/j.trsl.2021.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022]
Abstract
Plasma leakage is a hallmark process in dengue viral (DENV) infection that occurs due to the loss of vascular integrity in endothelial cells. Endoglin (ENG) and Syndecan-1 (SDC-1) are released by activated endothelial cells; however, the complete dynamics of its expression at the gene and protein levels during the course of DENV infection remains unknown. In the present study, we quantified the mRNA and soluble protein levels of ENG and SDC-1 in dengue cases during febrile, defervescence, and convalescence stages in Dengue without Warning Sign (DWOW-15), Dengue with Warning Sign (DWW-22), and Severe Dengue cases (SD-10) compared to nondengue Other Febrile Illness (OFI-10) and healthy control (HC-8). Respective protein and mRNA levels along with clinical characters were further analyzed for their efficacy in predicting disease outcomes using Support Vector Machine (SVM). We observed a steady and significant (P ≤ 0.01) increase in the levels of protein and mRNA of both the ENG and SDC-1 towards defervescence which is considered a critical phase in both severe and non-severe dengue cases. Importantly during the critical phase, the levels were significantly higher (P ≤ 0.001) in SD cases compared to DWW, DWOW, and OFI controls. However, at the time of admission (febrile), no such significant changes were observed within dengue, OFI, and healthy controls. SVM analysis revealed that the serum levels of ENG and SDC-1 along with other clinical symptoms could predict the disease severity with 100% accuracy. Based on the results we have proposed a mechanism on how ENG and SDC-1 could be involved in vascular dysfunction rather than just being a biomarker.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Central Inter-Disciplinary Research Facility (CIDRF), School of Biological Science, Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, India
| | - Shalinda Adikari
- Department of Information System and Analytics, National University of Singapore (NUS), Singapore, Republic of Singapore
| | - Lokesh Shanmugam
- Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, India
| | - Joshy M Easow
- Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, India
| | - Agieshkumar Balakrishna Pillai
- Central Inter-Disciplinary Research Facility (CIDRF), School of Biological Science, Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, India.
| |
Collapse
|
20
|
Contribution of Syndecans to the Cellular Entry of SARS-CoV-2. Int J Mol Sci 2021; 22:ijms22105336. [PMID: 34069441 PMCID: PMC8159090 DOI: 10.3390/ijms22105336] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 12/13/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel emerging pathogen causing an unprecedented pandemic in 21st century medicine. Due to the significant health and economic burden of the current SARS-CoV-2 outbreak, there is a huge unmet medical need for novel interventions effectively blocking SARS-CoV-2 infection. Unknown details of SARS-CoV-2 cellular biology hamper the development of potent and highly specific SARS-CoV-2 therapeutics. Angiotensin-converting enzyme-2 (ACE2) has been reported to be the primary receptor for SARS-CoV-2 cellular entry. However, emerging scientific evidence suggests the involvement of additional membrane proteins, such as heparan sulfate proteoglycans, in SARS-CoV-2 internalization. Here, we report that syndecans, the evolutionarily conserved family of transmembrane proteoglycans, facilitate the cellular entry of SARS-CoV-2. Among syndecans, the lung abundant syndecan-4 was the most efficient in mediating SARS-CoV-2 uptake. The S1 subunit of the SARS-CoV-2 spike protein plays a dominant role in the virus's interactions with syndecans. Besides the polyanionic heparan sulfate chains, other parts of the syndecan ectodomain, such as the cell-binding domain, also contribute to the interaction with SARS-CoV-2. During virus internalization, syndecans colocalize with ACE2, suggesting a jointly shared internalization pathway. Both ACE2 and syndecan inhibitors exhibited significant efficacy in reducing the cellular entry of SARS-CoV-2, thus supporting the complex nature of internalization. Data obtained on syndecan specific in vitro assays present syndecans as novel cellular targets of SARS-CoV-2 and offer molecularly precise yet simple strategies to overcome the complex nature of SARS-CoV-2 infection.
Collapse
|
21
|
Trybala E, Peerboom N, Adamiak B, Krzyzowska M, Liljeqvist JÅ, Bally M, Bergström T. Herpes Simplex Virus Type 2 Mucin-Like Glycoprotein mgG Promotes Virus Release from the Surface of Infected Cells. Viruses 2021; 13:v13050887. [PMID: 34065826 PMCID: PMC8150390 DOI: 10.3390/v13050887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 01/08/2023] Open
Abstract
The contribution of virus components to liberation of herpes simplex virus type 2 (HSV-2) progeny virions from the surface of infected cells is poorly understood. We report that the HSV-2 mutant deficient in the expression of a mucin-like membrane-associated glycoprotein G (mgG) exhibited defect in the release of progeny virions from infected cells manifested by ~2 orders of magnitude decreased amount of infectious virus in a culture medium as compared to native HSV-2. Electron microscopy revealed that the mgG deficient virions were produced in infected cells and present at the cell surface. These virions could be forcibly liberated to a nearly native HSV-2 level by the treatment of cells with glycosaminoglycan (GAG)-mimicking oligosaccharides. Comparative assessment of the interaction of mutant and native virions with surface-immobilized chondroitin sulfate GAG chains revealed that while the mutant virions associated with GAGs ~fourfold more extensively, the lateral mobility of bound virions was much poorer than that of native virions. These data indicate that the mgG of HSV-2 balances the virus interaction with GAG chains, a feature critical to prevent trapping of the progeny virions at the surface of infected cells.
Collapse
Affiliation(s)
- Edward Trybala
- Department of Infectious Diseases, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, SE-413 46 Göteborg, Sweden; (E.T.); (B.A.); (M.K.); (J.-Å.L.)
| | - Nadia Peerboom
- Department of Physics, Chalmers University of Technology, SE-412 96 Göteborg, Sweden;
| | - Beata Adamiak
- Department of Infectious Diseases, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, SE-413 46 Göteborg, Sweden; (E.T.); (B.A.); (M.K.); (J.-Å.L.)
| | - Malgorzata Krzyzowska
- Department of Infectious Diseases, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, SE-413 46 Göteborg, Sweden; (E.T.); (B.A.); (M.K.); (J.-Å.L.)
| | - Jan-Åke Liljeqvist
- Department of Infectious Diseases, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, SE-413 46 Göteborg, Sweden; (E.T.); (B.A.); (M.K.); (J.-Å.L.)
| | - Marta Bally
- Department of Clinical Microbiology, Umeå University, SE-901 85 Umeå, Sweden;
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-901 85 Umeå, Sweden
| | - Tomas Bergström
- Department of Infectious Diseases, Section for Clinical Virology, Institute of Biomedicine, University of Gothenburg, SE-413 46 Göteborg, Sweden; (E.T.); (B.A.); (M.K.); (J.-Å.L.)
- Correspondence:
| |
Collapse
|
22
|
Li Y, Liu D, Wang Y, Su W, Liu G, Dong W. The Importance of Glycans of Viral and Host Proteins in Enveloped Virus Infection. Front Immunol 2021; 12:638573. [PMID: 33995356 PMCID: PMC8116741 DOI: 10.3389/fimmu.2021.638573] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Animal viruses are parasites of animal cells that have characteristics such as heredity and replication. Viruses can be divided into non-enveloped and enveloped viruses if a lipid bilayer membrane surrounds them or not. All the membrane proteins of enveloped viruses that function in attachment to target cells or membrane fusion are modified by glycosylation. Glycosylation is one of the most common post-translational modifications of proteins and plays an important role in many biological behaviors, such as protein folding and stabilization, virus attachment to target cell receptors and inhibition of antibody neutralization. Glycans of the host receptors can also regulate the attachment of the viruses and then influence the virus entry. With the development of glycosylation research technology, the research and development of novel virus vaccines and antiviral drugs based on glycan have received increasing attention. Here, we review the effects of host glycans and viral proteins on biological behaviors of viruses, and the opportunities for prevention and treatment of viral infectious diseases.
Collapse
Affiliation(s)
- Yuqing Li
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Dongqi Liu
- The Queen's University of Belfast Joint College, China Medical University, Shenyang, China
| | - Yating Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Wenquan Su
- Dalian Medical University, Dalian, China
| | - Gang Liu
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Weijie Dong
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| |
Collapse
|
23
|
Antiviral strategies should focus on stimulating the biosynthesis of heparan sulfates, not their inhibition. Life Sci 2021; 277:119508. [PMID: 33865880 PMCID: PMC8046744 DOI: 10.1016/j.lfs.2021.119508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/23/2022]
Abstract
Antiviral strategies for viruses that utilize proteoglycan core proteins (syndecans and glypicans) as receptors should focus on heparan sulfate (HS) biosynthesis rather than on inhibition of these sugar chains. Here, we show that heparin and certain xylosides, which exhibit in vitro viral entry inhibitory properties against HSV-1, HSV-2, HPV-16, HPV-31, HVB, HVC, HIV-1, HTLV-1, SARS-CoV-2, HCMV, DENV-1, and DENV-2, stimulated HS biosynthesis at the cell surface 2- to 3-fold for heparin and up to 10-fold for such xylosides. This is consistent with the hypothesis from a previous study that for core protein attachment, viruses are glycosylated at HS attachment sites (i.e., serine residues intended to receive the D-xylose molecule for initiating HS chains). Heparanase overexpression, endocytic entry, and syndecan shedding enhancement, all of which are observed during viral infection, lead to glycocalyx deregulation and appear to be direct consequences of this hypothesis. In addition to the appearance of type 2 diabetes and the degradation of HS observed during viral infection, we linked this hypothesis to that proposed in a previous publication.
Collapse
|
24
|
Koganti R, Memon A, Shukla D. Emerging Roles of Heparan Sulfate Proteoglycans in Viral Pathogenesis. Semin Thromb Hemost 2021; 47:283-294. [PMID: 33851373 DOI: 10.1055/s-0041-1725068] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Heparan sulfate is a glycosaminoglycan present in nearly all mammalian tissues. Heparan sulfate moieties are attached to the cell surface via heparan sulfate proteoglycans (HSPGs) which are composed of a protein core bound to multiple heparan sulfate chains. HSPGs contribute to the structural integrity of the extracellular matrix and participate in cell signaling by releasing bound cytokines and chemokines once cleaved by an enzyme, heparanase. HSPGs are often exploited by viruses during infection, particularly during attachment and egress. Loss or inhibition of HSPGs initially during infection can yield significant decreases in viral entry and infectivity. In this review, we provide an overview of HSPGs in the lifecycle of multiple viruses, including herpesviruses, human immunodeficiency virus, dengue virus, human papillomavirus, and coronaviruses.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Abdullah Memon
- College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois.,Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
25
|
Vlodavsky I, Barash U, Nguyen HM, Yang SM, Ilan N. Biology of the Heparanase-Heparan Sulfate Axis and Its Role in Disease Pathogenesis. Semin Thromb Hemost 2021; 47:240-253. [PMID: 33794549 DOI: 10.1055/s-0041-1725066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell surface proteoglycans are important constituents of the glycocalyx and participate in cell-cell and cell-extracellular matrix (ECM) interactions, enzyme activation and inhibition, and multiple signaling routes, thereby regulating cell proliferation, survival, adhesion, migration, and differentiation. Heparanase, the sole mammalian heparan sulfate degrading endoglycosidase, acts as an "activator" of HS proteoglycans, thus regulating tissue hemostasis. Heparanase is a multifaceted enzyme that together with heparan sulfate, primarily syndecan-1, drives signal transduction, immune cell activation, exosome formation, autophagy, and gene transcription via enzymatic and nonenzymatic activities. An important feature is the ability of heparanase to stimulate syndecan-1 shedding, thereby impacting cell behavior both locally and distally from its cell of origin. Heparanase releases a myriad of HS-bound growth factors, cytokines, and chemokines that are sequestered by heparan sulfate in the glycocalyx and ECM. Collectively, the heparan sulfate-heparanase axis plays pivotal roles in creating a permissive environment for cell proliferation, differentiation, and function, often resulting in the pathogenesis of diseases such as cancer, inflammation, endotheliitis, kidney dysfunction, tissue fibrosis, and viral infection.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Uri Barash
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
26
|
Syntenin-knock out reduces exosome turnover and viral transduction. Sci Rep 2021; 11:4083. [PMID: 33602969 PMCID: PMC7892569 DOI: 10.1038/s41598-021-81697-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/23/2020] [Indexed: 12/19/2022] Open
Abstract
Exosomal transfers represent an important mode of intercellular communication. Syntenin is a small scaffold protein that, when binding ALIX, can direct endocytosed syndecans and syndecan cargo to budding endosomal membranes, supporting the formation of intraluminal vesicles that compose the source of a major class of exosomes. Syntenin, however, can also support the recycling of these same components to the cell surface. Here, by studying mice and cells with syntenin-knock out, we identify syntenin as part of dedicated machinery that integrates both the production and the uptake of secreted vesicles, supporting viral/exosomal exchanges. This study significantly extends the emerging role of heparan sulfate proteoglycans and syntenin as key components for macromolecular cargo internalization into cells.
Collapse
|
27
|
Gopal S, Arokiasamy S, Pataki C, Whiteford JR, Couchman JR. Syndecan receptors: pericellular regulators in development and inflammatory disease. Open Biol 2021; 11:200377. [PMID: 33561383 PMCID: PMC8061687 DOI: 10.1098/rsob.200377] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
The syndecans are the major family of transmembrane proteoglycans, usually bearing multiple heparan sulfate chains. They are present on virtually all nucleated cells of vertebrates and are also present in invertebrates, indicative of a long evolutionary history. Genetic models in both vertebrates and invertebrates have shown that syndecans link to the actin cytoskeleton and can fine-tune cell adhesion, migration, junction formation, polarity and differentiation. Although often associated as co-receptors with other classes of receptors (e.g. integrins, growth factor and morphogen receptors), syndecans can nonetheless signal to the cytoplasm in discrete ways. Syndecan expression levels are upregulated in development, tissue repair and an array of human diseases, which has led to the increased appreciation that they may be important in pathogenesis not only as diagnostic or prognostic agents, but also as potential targets. Here, their functions in development and inflammatory diseases are summarized, including their potential roles as conduits for viral pathogen entry into cells.
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 3800, Australia
| | - Samantha Arokiasamy
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Csilla Pataki
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - James R. Whiteford
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - John R. Couchman
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
28
|
Rangarajan S, Richter JR, Richter RP, Bandari SK, Tripathi K, Vlodavsky I, Sanderson RD. Heparanase-enhanced Shedding of Syndecan-1 and Its Role in Driving Disease Pathogenesis and Progression. J Histochem Cytochem 2020; 68:823-840. [PMID: 32623935 PMCID: PMC7711244 DOI: 10.1369/0022155420937087] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/29/2020] [Indexed: 02/08/2023] Open
Abstract
Both heparanase and syndecan-1 are known to be present and active in disease pathobiology. An important feature of syndecan-1 related to its role in pathologies is that it can be shed from the surface of cells as an intact ectodomain composed of the extracellular core protein and attached heparan sulfate and chondroitin sulfate chains. Shed syndecan-1 remains functional and impacts cell behavior both locally and distally from its cell of origin. Shedding of syndecan-1 is initiated by a variety of stimuli and accomplished predominantly by the action of matrix metalloproteinases. The accessibility of these proteases to the core protein of syndecan-1 is enhanced, and shedding facilitated, when the heparan sulfate chains of syndecan-1 have been shortened by the enzymatic activity of heparanase. Interestingly, heparanase also enhances shedding by upregulating the expression of matrix metalloproteinases. Recent studies have revealed that heparanase-induced syndecan-1 shedding contributes to the pathogenesis and progression of cancer and viral infection, as well as other septic and non-septic inflammatory states. This review discusses the heparanase/shed syndecan-1 axis in disease pathogenesis and progression, the potential of targeting this axis therapeutically, and the possibility that this axis is widespread and of influence in many diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Israel Vlodavsky
- The University of Alabama at Birmingham, Birmingham, Alabama, and Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
29
|
Kouwenberg M, Rops A, Bakker-van Bebber M, Diepeveen L, Götte M, Hilbrands L, van der Vlag J. Role of syndecan-1 in the interaction between dendritic cells and T cells. PLoS One 2020; 15:e0230835. [PMID: 32701966 PMCID: PMC7377417 DOI: 10.1371/journal.pone.0230835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/01/2020] [Indexed: 01/19/2023] Open
Abstract
Syndecan-1 (Sdc-1) is a heparan sulfate proteoglycan that can bind cytokines and chemokines via its heparan sulfate side chains, and has immunomodulatory properties in experimental models. Sdc-1 expression has been reported on dendritic cells (DC) and T cells. The potential role of Sdc-1 in DC-T cell interaction has not been investigated yet. We postulate that Sdc-1 is involved in DC-T cell interaction and may influence graft survival in an allogeneic transplant model. Sdc-1 expression on bone marrow-derived DC and T cells was analyzed by flow cytometry. Unstimulated and LPS stimulated Sdc-1 deficient DC were evaluated in vitro for phenotype and stimulatory capacity in mixed lymphocyte reaction. Sdc-1 deficient T cells were evaluated for proliferative capacity and differentiation in a mixed lymphocyte reaction and a proliferation assay. Allograft survival was evaluated in a fully MHC mismatched heterotopic heart transplant model, with either Sdc-1 deficient donors or recipients. Sdc-1 was expressed on the cell surface of unstimulated and LPS matured DC. Sdc-1 deficiency had no effect on expression of co-stimulatory molecules, cytokine production or T cell stimulatory capacity as compared to WT DC. Sdc-1 expression was not detectable on WT T cells, although intracellular Sdc-1 expression could be demonstrated after ConA activation. Sdc-1 deficient T cells showed reduced proliferation upon DC or ConA stimulation and reduced IL-17 production upon ConA stimulation, compared to WT T cells. Sdc-1 deficiency of either allograft or recipient did not prolong allograft survival. In conclusion, Sdc-1 is expressed on the cell surface of DC, where its absence does not affect DC phenotype or T cell stimulatory capacity. Sdc-1 is intracellularly expressed in ConA activated T cells. Sdc-1 deficiency in T cells results in a reduced proliferative response in vitro, as induced by DC and ConA. Sdc-1 deficiency in donor or recipient does not affect allograft survival.
Collapse
Affiliation(s)
- M. Kouwenberg
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A. Rops
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M. Bakker-van Bebber
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L. Diepeveen
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M. Götte
- Department of Gynecology and Obstetrics, University of Münster, Münster, Germany
| | - L. Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J. van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
30
|
Zhao T, Cui L, Yu X, Zhang Z, Chen Q, Hua X. Proteome Analysis Reveals Syndecan 1 Regulates Porcine Sapelovirus Replication. Int J Mol Sci 2020; 21:E4386. [PMID: 32575635 PMCID: PMC7352226 DOI: 10.3390/ijms21124386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/03/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
Porcine sapelovirus A (PSV) is a single stranded, positive-sense, non-enveloped RNA virus that causes enteritis, pneumonia, polioencephalomyelitis, and reproductive disorders in pigs. Research on PSV infection and interaction with host cells is unclear. In this study, we applied tandem mass tag proteomics analysis to investigate the differentially expressed proteins (DEPs) in PSV-infected pig kidney (PK)-15 cells and explored the interactions between PSV and host cells. Here we mapped 181 DEPs, including 59 up-regulated and 122 down-regulated DEPs. Among them, osteopontin (SPP1), induced protein with tetratricopeptide repeats 5 (IFIT5), ISG15 ubiquitin-like modifier (ISG15), vinculin (VCL), and syndecan-1 (SDC1) were verified significantly changed using RT-qPCR. Additionally, overexpression of SDC1 promoted PSV viral protein (VP)1 synthesis and virus titer, and silencing of SDC1 revealed the opposite results. Our findings show that SDC1 is a novel host protein and plays crucial roles in regulating PSV replication.
Collapse
Affiliation(s)
- Tingting Zhao
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (T.Z.); (L.C.)
| | - Li Cui
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (T.Z.); (L.C.)
| | - Xiangqian Yu
- Shanghai Pudong New Area Center for Animal Disease Control and Prevention, Shanghai 200136, China; (X.Y.); (Z.Z.)
| | - Zhonghai Zhang
- Shanghai Pudong New Area Center for Animal Disease Control and Prevention, Shanghai 200136, China; (X.Y.); (Z.Z.)
| | - Qi Chen
- Shanghai Animal Disease Control Center, Shanghai 201103, China;
| | - Xiuguo Hua
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (T.Z.); (L.C.)
| |
Collapse
|
31
|
Wang L, Wang R, Xu C, Zhou H. Pathogenesis of Herpes Stromal Keratitis: Immune Inflammatory Response Mediated by Inflammatory Regulators. Front Immunol 2020; 11:766. [PMID: 32477330 PMCID: PMC7237736 DOI: 10.3389/fimmu.2020.00766] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022] Open
Abstract
Herpes stromal keratitis (HSK) is one of the primary diseases that cause vision loss or even blindness after herpes simplex virus (HSV)-1 infection. HSK-associated vision impairment is predominantly due to corneal scarring and neovascularization caused by inflammation. In the infected cornea, HSV can activate innate and adaptive immune responses of host cells, which triggers a cascade of reactions that leads to the release of inflammatory cytokines, chemokines, microRNA, and other regulatory factors that have stimulating or inhibitory effects on tissue. Physiologically, host cells show homeostasis. In this review, we summarize the factors involved in HSK pathogenesis from the perspective of immunity, molecules, and pathological angiogenesis. We also describe in detail the pathogenesis of chronic inflammatory lesions of the corneal stroma in response to HSV-1 infection.
Collapse
Affiliation(s)
- Li Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China.,Department of Ophthalmology, Jilin City Central Hospital, Jilin, China
| | - Runbiao Wang
- Department of Ophthalmology, Jilin City Central Hospital, Jilin, China
| | - Chuyang Xu
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hongyan Zhou
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Dai X, Zhang X, Ostrikov K, Abrahamyan L. Host receptors: the key to establishing cells with broad viral tropism for vaccine production. Crit Rev Microbiol 2020; 46:147-168. [PMID: 32202955 PMCID: PMC7113910 DOI: 10.1080/1040841x.2020.1735992] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell culture-based vaccine technology is a flexible and convenient approach for vaccine production that requires adaptation of the vaccine strains to the new cells. Driven by the motivation to develop a broadly permissive cell line for infection with a wide range of viruses, we identified a set of the most relevant host receptors involved in viral attachment and entry. This identification was done through a review of different viral entry pathways and host cell lines, and in the context of the Baltimore classification of viruses. In addition, we indicated the potential technical problems and proposed some solutions regarding how to modify the host cell genome in order to meet industrial requirements for mass production of antiviral vaccines. Our work contributes to a finer understanding of the importance of breaking the host–virus recognition specificities for the possibility of creating a cell line feasible for the production of vaccines against a broad spectrum of viruses.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xuanhao Zhang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Kostya Ostrikov
- School of Chemistry and Physics and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Levon Abrahamyan
- Faculty of Veterinary Medicine, Swine and Poultry Infectious Diseases Research Center (CRIPA), Research Group on Infectious Diseases in Production Animals (GREMIP), Université de Montréal, Saint-Hyacinthe, Canada
| |
Collapse
|
33
|
Heparanase-Regulated Syndecan-1 Shedding Facilitates Herpes Simplex Virus 1 Egress. J Virol 2020; 94:JVI.01672-19. [PMID: 31827001 DOI: 10.1128/jvi.01672-19] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) can infect virtually all cell types in vitro An important reason lies in its ability to exploit heparan sulfate (HS) for attachment to cells. HS is a ubiquitous glycosaminoglycan located on the cell surface and tethered to proteoglycans such as syndecan-1. Previously, we have shown that heparanase (HPSE) facilitates the release of viral particles by cleaving HS. Here, we demonstrate that HPSE is a master regulator where, in addition to directly enabling viral release via HS removal, it also facilitates cleavage of HS-containing ectodomains of syndecan-1, thereby further enhancing HSV-1 egress from infected cells. Syndecan-1 cleavage is mediated by upregulation of matrix metalloproteases (MMPs) that accompanies higher HPSE expression in infected cells. By overexpressing HPSE, we have identified MMP-3 and MMP-7 as important sheddases of syndecan-1 shedding in corneal epithelial cells, which are natural targets of HSV-1 infection. MMP-3 and MMP-7 were also naturally upregulated during HSV-1 infection. Altogether, this paper shows a new connection between HSV-1 release and syndecan-1 shedding, a phenomenon that is regulated by HPSE and executed by the MMPs. Our results also identify new molecular markers for HSV-1 infection and new targets for future interventions.IMPORTANCE HSV-1 is a common cause of recurrent viral infections in humans. The virus can cause a range of mucosal pathologies. Efficient viral egress from infected cells is an important step for HSV-1 transmission and virus-associated pathologies. Host mechanisms that contribute to HSV-1 egress from infected cells are poorly understood. Syndecan-1 is a common heparan sulfate proteoglycan expressed by many natural target cells. Despite its known connection with heparanase, a recently identified mediator of HSV-1 release, syndecan-1 has not been previously investigated in HSV-1 release. In this study, we demonstrate that the shedding of syndecan-1 by MMP-3 and MMP-7 supports viral egress. We show that the mechanism behind the activation of these MMPs is mediated by heparanase, which is upregulated upon HSV-1 infection. Our study elucidates a new connection between HSV-1 egress, heparanase, and matrix metallopeptidases; identifies new molecular markers of infection; and provides potential new targets for therapeutic interventions.
Collapse
|
34
|
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
|
35
|
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: 0.8] [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
|
36
|
Holmes VM, Maluquer de Motes C, Richards PT, Roldan J, Bhargava AK, Orange JS, Krummenacher C. Interaction between nectin-1 and the human natural killer cell receptor CD96. PLoS One 2019; 14:e0212443. [PMID: 30759143 PMCID: PMC6373967 DOI: 10.1371/journal.pone.0212443] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/01/2019] [Indexed: 12/17/2022] Open
Abstract
Regulation of Natural Killer (NK) cell activity is achieved by the integration of both activating and inhibitory signals acquired at the immunological synapse with potential target cells. NK cells express paired receptors from the immunoglobulin family which share common ligands from the nectin family of adhesion molecules. The activating receptor CD226 (DNAM-1) binds to nectin-2 and CD155, which are also recognized by the inhibitory receptor TIGIT. The third receptor in this family is CD96, which is less well characterized and may have different functions in human and mouse models. Human CD96 interacts with CD155 and ligation of this receptor activates NK cells, while in mice the presence of CD96 correlates with decreased NK cell activation. Mouse CD96 also binds nectin-1, but the effect of this interaction has not yet been determined. Here we show that human nectin-1 directly interacts with CD96 in vitro. The binding site for CD96 is located on the nectin-1 V-domain, which comprises a canonical interface that is shared by nectins to promote cell adhesion. The affinity of nectin-1 for CD96 is lower than for other nectins such as nectin-3 and nectin-1 itself. However, the affinity of nectin-1 for CD96 is similar to its affinity for herpes simplex virus glycoprotein D (HSV gD), which binds the nectin-1 V-domain during virus entry. The affinity of human CD96 for nectin-1 is lower than for its known activating ligand CD155. We also found that human erythroleukemia K562 cells, which are commonly used as susceptible targets to assess NK cell cytotoxicity did not express nectin-1 on their surface and were resistant to HSV infection. When expressed in K562 cells, nectin-1-GFP accumulated at cell contacts and allowed HSV entry. Furthermore, overexpression of nectin-1-GFP led to an increased susceptibility of K562 cells to NK-92 cell cytotoxicity.
Collapse
Affiliation(s)
- Veronica M. Holmes
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Paige T. Richards
- Department of Biological Sciences, Rowan University, Glassboro, New Jersey, United States of America
| | - Jessenia Roldan
- Department of Biological Sciences, Rowan University, Glassboro, New Jersey, United States of America
| | - Arjun K. Bhargava
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jordan S. Orange
- Department of Pediatrics, Columbia University, New York, New York, United States of America
| | - Claude Krummenacher
- Department of Biological Sciences, Rowan University, Glassboro, New Jersey, United States of America
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, New Jersey, United States of America
| |
Collapse
|
37
|
Perkins TN, Peeters PM, Albrecht C, Schins RPF, Dentener MA, Mossman BT, Wouters EFM, Reynaert NL. Crystalline silica alters Sulfatase-1 expression in rat lungs which influences hyper-proliferative and fibrogenic effects in human lung epithelial cells. Toxicol Appl Pharmacol 2018; 348:43-53. [PMID: 29673857 DOI: 10.1016/j.taap.2018.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/06/2018] [Accepted: 04/10/2018] [Indexed: 12/27/2022]
Abstract
Lung epithelial cells are the first cell-type to come in contact with hazardous dust materials. Upon deposition, they invoke complex reactions in attempt to eradicate particles from the airways, and repair damage. The cell surface is composed of a heterogeneous network of matrix proteins and proteoglycans, which act as scaffold and control cell-signaling networks. These functions are controlled, in part, by the sulfation patterns of heparin-sulfate proteoglycans (HSPGs), which are enzymatically regulated. Although there is evidence of altered HSPG-sulfation in idiopathic pulmonary fibrosis (IPF), this is not investigated in silicosis. Our previous studies revealed down-regulation of Sulfatase-1 (SULF1) in human bronchial epithelial cells (BECs) by crystalline silica (CS). In this study, CS-induced down-regulation of SULF1, and increases in Sulfated-HSPGs, were determined in human BECs, and in rat lungs. By siRNA and plasmid transfection techniques the effects of SULF1 expression on silica-induced fibrogenic and proliferative gene expression were determined. These studies confirmed down-regulation of SULF1 and subsequent increases in sulfated-HSPGs in vitro. Moreover, short-term exposure of rats to CS resulted in similar changes in vivo. Conversely, effects were reversed after long term CS exposure of rats. SULF1 knockdown, and overexpression alleviated and exacerbated silica-induced decrease in cell viability, respectively. Furthermore, overexpression of SULF1 promoted silica-induced proliferative and fibrogenic gene expression, and collagen production. These findings demonstrate that the HSPG modification enzyme SULF1 and HSPG sulfation are altered by CS in vitro and in vivo. Furthermore, these changes may contribute to CS-induced lung pathogenicity by affecting injury tolerance, hyperproliferation, and fibrotic effects.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Respiratory Medicine, Maastricht University, Medical Centre, Maastricht, The Netherlands; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Paul M Peeters
- Department of Respiratory Medicine, Maastricht University, Medical Centre, Maastricht, The Netherlands; IUF, Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Catrin Albrecht
- IUF, Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Roel P F Schins
- IUF, Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Mieke A Dentener
- Department of Respiratory Medicine, Maastricht University, Medical Centre, Maastricht, The Netherlands.
| | - Brooke T Mossman
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA.
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University, Medical Centre, Maastricht, The Netherlands.
| | - Niki L Reynaert
- Department of Respiratory Medicine, Maastricht University, Medical Centre, Maastricht, The Netherlands.
| |
Collapse
|
38
|
Sobhy H. A comparative review of viral entry and attachment during large and giant dsDNA virus infections. Arch Virol 2017; 162:3567-3585. [PMID: 28866775 PMCID: PMC5671522 DOI: 10.1007/s00705-017-3497-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/13/2017] [Indexed: 12/19/2022]
Abstract
Viruses enter host cells via several mechanisms, including endocytosis, macropinocytosis, and phagocytosis. They can also fuse at the plasma membrane and can spread within the host via cell-to-cell fusion or syncytia. The mechanism used by a given viral strain depends on its external topology and proteome and the type of cell being entered. This comparative review discusses the cellular attachment receptors and entry pathways of dsDNA viruses belonging to the families Adenoviridae, Baculoviridae, Herpesviridae and nucleocytoplasmic large DNA viruses (NCLDVs) belonging to the families Ascoviridae, Asfarviridae, Iridoviridae, Phycodnaviridae, and Poxviridae, and giant viruses belonging to the families Mimiviridae and Marseilleviridae as well as the proposed families Pandoraviridae and Pithoviridae. Although these viruses have several common features (e.g., topology, replication and protein sequence similarities) they utilize different entry pathways to infect wide-range of hosts, including humans, other mammals, invertebrates, fish, protozoa and algae. Similarities and differences between the entry methods used by these virus families are highlighted, with particular emphasis on viral topology and proteins that mediate viral attachment and entry. Cell types that are frequently used to study viral entry are also reviewed, along with other factors that affect virus-host cell interactions.
Collapse
Affiliation(s)
- Haitham Sobhy
- Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden.
| |
Collapse
|
39
|
Heparanase Upregulation Contributes to Porcine Reproductive and Respiratory Syndrome Virus Release. J Virol 2017; 91:JVI.00625-17. [PMID: 28490587 DOI: 10.1128/jvi.00625-17] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/02/2017] [Indexed: 12/27/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) continues to cause substantial economic losses to the pig industry worldwide. Heparan sulfate (HS) is used by PRRSV for initial attachment to target cells. However, the role of HS in the late phase of PRRSV infection and the mechanism of virus release from host cells remain largely unknown. In this study, we showed that PRRSV infection caused a decrease in HS expression and upregulated heparanase, the only known enzyme capable of degrading HS. We subsequently demonstrated that the NF-κB signaling pathway and cathepsin L protease were involved in regulation of PRRSV infection-induced heparanase. In addition, we found that ablation of heparanase expression using small interfering RNA duplexes increased cell surface expression of HS and suppressed PRRSV replication and release, whereas overexpression of heparanase reduced HS surface expression and enhanced PRRSV replication and release. These data suggest that PRRSV activates NF-κB and cathepsin L to upregulate and process heparanase, and then the active heparanase cleaves HS, resulting in viral release. Our findings provide new insight into the molecular mechanism of PRRSV egress from host cells, which might help us to further understand PRRSV pathogenesis.IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV) causes great economic losses each year to the pig industry worldwide. The molecular mechanism of PRRSV release from host cells largely remains a mystery. In this study, we demonstrate that PRRSV activates NF-κB and cathepsin L to upregulate and process heparanase, and then the active heparanase is released to the extracellular space and exerts enzymatic activity to cleave heparan sulfate, resulting in viral release. Our findings provide new insight into the molecular mechanism of PRRSV egress from host cells, which might help us to further understand PRRSV pathogenesis.
Collapse
|
40
|
Induction of Multiple miR-200/182 Members in the Brains of Mice Are Associated with Acute Herpes Simplex Virus 1 Encephalitis. PLoS One 2017; 12:e0169081. [PMID: 28045967 PMCID: PMC5207681 DOI: 10.1371/journal.pone.0169081] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Important roles of microRNAs (miRNAs) in regulating the host response during viral infection have begun to be defined. However, little is known about the functional roles of miRNAs within an in vivo acute viral encephalitis model. We therefore identified global changes in miRNA expression during acute herpes simplex virus type 1 (HSV-1) encephalitis (HSVE) in mice. We found that many of the highly upregulated miRNAs (miR-155, miR-146a and miR-15b) detected in HSV-1 infected brain tissue are known regulators of inflammation and innate immunity. We also observed upregulation of 7 members belonging to the related group of miRNAs, the miR-200 family and miR-182 cluster (miR-200/182). Using in situ hybridization, we found that these miRNAs co-localized to regions of the brain with severe HSVE-related pathology and were upregulated in various cell types including neurons. Induction was apparent but not limited to cells in which HSV-1 was detected by immunohistochemistry, suggesting possible roles of these miRNAs in the host response to viral-induced tissue damage. Bioinformatic prediction combined with gene expression profiling revealed that the induced miR-200/182 members could regulate the biosynthesis of heparan sulfate proteoglycans. Using luciferase assays, we found that miR-96, miR-141, miR-183 and miR-200c all potentially targeted the syndecan-2 gene (Sdc2), which codes for a cell surface heparan sulfate proteoglycan involved in HSV-1 cellular attachment and entry.
Collapse
|
41
|
Tsidulko AY, Matskova L, Astakhova LA, Ernberg I, Grigorieva EV. Proteoglycan expression correlates with the phenotype of malignant and non-malignant EBV-positive B-cell lines. Oncotarget 2016; 6:43529-39. [PMID: 26527314 PMCID: PMC4791248 DOI: 10.18632/oncotarget.5984] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/06/2015] [Indexed: 11/25/2022] Open
Abstract
The involvement of proteoglycans (PGs) in EBV-host interactions and lymphomagenesis remains poorly investigated. In this study, expression of major proteoglycans (syndecan-1, glypican-1, perlecan, versican, brevican, aggrecan, NG2, serglycin, decorin, biglycan, lumican, CD44), heparan sulphate (HS) metabolic system (EXT1/2, NDST1/2, GLCE, HS2ST1, HS3ST1/2, HS6ST1/2, SULF1/2, HPSE) and extracellular matrix (ECM) components (collagen 1A1, fibronectin, elastin) in primary B cells and EBV carrying cell lines with different phenotypes, patterns of EBV-host cell interaction and viral latency stages (type I-III) was investigated. Primary B cells expressed a wide repertoire of PGs (dominated by serglycin and CD44) and ECM components. Lymphoblastoid EBV+ B cell lines (LCLs) showed specific PG expression with down-regulation of CD44 and ECM components and up-regulation of serglycin and perlecan/HSPG2. For Burkitt's lymphoma cells (BL), serglycin was down-regulated in BL type III cells and perlecan in type I BL cells. The biosynthetic machinery for HS was active in all cell lines, with some tendency to be down-regulated in BL cells. 5'-aza-dC and/or Trichostatin A resulted in transcriptional upregulation of the genes, suggesting that low expression of ECM components, proteoglycan core proteins and HS biosynthetic system is due to epigenetic suppression in type I cells. Taken together, our data show that proteoglycans are expressed in primary B lymphocytes whereas they are not or only partly expressed in EBV-carrying cell lines, depending on their latency type program.
Collapse
Affiliation(s)
- Alexandra Y Tsidulko
- Institute of Molecular Biology and Biophysics, Novosibirsk, Russia.,Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Liudmila Matskova
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | | | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Elvira V Grigorieva
- Institute of Molecular Biology and Biophysics, Novosibirsk, Russia.,Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
42
|
Waters AM, Stafman LL, Garner EF, Mruthyunjayappa S, Stewart JE, Friedman GK, Coleman JM, Markert JM, Gillespie GY, Beierle EA. Effect of Repeat Dosing of Engineered Oncolytic Herpes Simplex Virus on Preclinical Models of Rhabdomyosarcoma. Transl Oncol 2016; 9:419-430. [PMID: 27751346 PMCID: PMC5067929 DOI: 10.1016/j.tranon.2016.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 12/16/2022] Open
Abstract
Rhabdomyosarcoma (RMS), a tumor of skeletal muscle origin, is the most common sarcoma of childhood. Despite multidrug chemotherapy regimens, surgical intervention, and radiation treatment, outcomes remain poor, especially in advanced disease, and novel therapies are needed for the treatment of these aggressive malignancies. Genetically engineered oncolytic viruses, such as herpes simplex virus-1 (HSV), are currently being explored as treatments for pediatric tumors. M002, an oncolytic HSV, has both copies of the γ134.5 gene deleted, enabling replication in tumor cells but thwarting infection of normal, postmitotic cells. We hypothesized that M002 would infect human RMS tumor cells and lead to decreased tumor cell survival in vitro and impede tumor growth in vivo. In the current study, we demonstrated that M002 could infect, replicate in, and decrease cell survival in both embryonal (ERMS) and alveolar rhabdomyosarcoma (ARMS) cells. Additionally, M002 reduced xenograft tumor growth and increased animal survival in both ARMS and ERMS. Most importantly, we showed for the first time that repeated dosing of oncolytic virus coupled with low-dose radiation provided improved tumor response in RMS. These findings provide support for the clinical investigation of oncolytic HSV in pediatric RMS.
Collapse
Affiliation(s)
- Alicia M Waters
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Laura L Stafman
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Evan F Garner
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Smitha Mruthyunjayappa
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Jerry E Stewart
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Gregory K Friedman
- Department of Pediatrics, Division of Hematology/Oncology, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Jennifer M Coleman
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - James M Markert
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - G Yancey Gillespie
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Elizabeth A Beierle
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233.
| |
Collapse
|
43
|
Zimmermann N, Saiga H, Houthuys E, Moura-Alves P, Koehler A, Bandermann S, Dorhoi A, Kaufmann SHE. Syndecans promote mycobacterial internalization by lung epithelial cells. Cell Microbiol 2016; 18:1846-1856. [PMID: 27279134 DOI: 10.1111/cmi.12627] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/11/2016] [Accepted: 06/05/2016] [Indexed: 01/16/2023]
Abstract
Pulmonary tuberculosis (TB) is an airborne disease caused by the intracellular bacterial pathogen Mycobacterium tuberculosis (Mtb). Alveolar epithelial cells and macrophages are the first point of contact for Mtb in the respiratory tract. However, the mechanisms of mycobacterial attachment to, and internalization by, nonprofessional phagocytes, such as epithelial cells, remain incompletely understood. We identified syndecan 4 (Sdc4) as mycobacterial attachment receptor on alveolar epithelial cells. Sdc4 mRNA expression was increased in human and mouse alveolar epithelial cells after mycobacterial infection. Sdc4 knockdown in alveolar epithelial cells or blocking with anti-Sdc4 antibody reduced mycobacterial attachment and internalization. At the molecular level, interactions between epithelial cells and mycobacteria involved host Sdc and the mycobacterial heparin-binding hemagglutinin adhesin. In vivo, Sdc1/Sdc4 double-knockout mice were more resistant to Mtb colonization of the lung. Our work reveals a role for distinct Sdcs in promoting mycobacterial entry into alveolar epithelial cells with impact on outcome of TB disease.
Collapse
Affiliation(s)
- Natalie Zimmermann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.,Research Group of Molecular Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hiroyuki Saiga
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Erica Houthuys
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Pedro Moura-Alves
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anne Koehler
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Silke Bandermann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anca Dorhoi
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
44
|
Skeate JG, Porras TB, Woodham AW, Jang JK, Taylor JR, Brand HE, Kelly TJ, Jung JU, Da Silva DM, Yuan W, Martin Kast W. Herpes simplex virus downregulation of secretory leukocyte protease inhibitor enhances human papillomavirus type 16 infection. J Gen Virol 2015; 97:422-434. [PMID: 26555393 DOI: 10.1099/jgv.0.000341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Herpes simplex virus (HSV) was originally implicated in the aetiology of cervical cancer, and although high-risk human papillomavirus (HPV) is now the accepted causative agent, the epidemiological link between HSV and HPV-associated cancers persists. The annexin A2 heterotetramer (A2t) has been shown to mediate infectious HPV type 16 (HPV16) uptake by human keratinocytes, and secretory leukocyte protease inhibitor (SLPI), an endogenous A2t ligand, inhibits HPV16 uptake and infection. Interestingly, HSV infection induces a sustained downregulation of SLPI in epithelial cells, which we hypothesized promotes HPV16 infection through A2t. Here, we show that in vitro infection of human keratinocytes with HSV-1 or HSV-2, but not with an HSV-1 ICP4 deletion mutant that does not downregulate SLPI, leads to a >70% reduction of SLPI mRNA and a >60% decrease in secreted SLPI protein. Consequently, we observed a significant increase in the uptake of HPV16 virus-like particles and gene transduction by HPV16 pseudovirions (two- and 2.5-fold, respectively) in HSV-1- and HSV-2-infected human keratinocyte cell cultures compared with uninfected cells, whereas exogenously added SLPI reversed this effect. Using a SiMPull (single-molecule pulldown) assay, we demonstrated that endogenously secreted SLPI interacts with A2t on epithelial cells in an autocrine/paracrine manner. These results suggested that ongoing HSV infection and resultant downregulation of local levels of SLPI may impart a greater susceptibility for keratinocytes to HPV16 infection through the host cell receptor A2t, providing a mechanism that may, in part, provide an explanation for the aetiological link between HSV and HPV-associated cancers.
Collapse
Affiliation(s)
- Joseph G Skeate
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Tania B Porras
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Andrew W Woodham
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Julie K Jang
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA
| | - Julia R Taylor
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Heike E Brand
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Thomas J Kelly
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Jae U Jung
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Diane M Da Silva
- Obstetrics & Gynecology, University of Southern California, Los Angeles, CA 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Weiming Yuan
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - W Martin Kast
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.,Obstetrics & Gynecology, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
45
|
Abstract
This paper discusses physical and structural aspects of the mechanisms herpes simplex virus (HSV) uses for membrane fusion. Calculations show that herpes simplex virus glycoprotein D has such avidity for its receptors that it can hold the virion against the plasma membrane of a neuron strongly enough for glycoprotein B (gB) to disrupt both leaflets of the bilayer. The strong electric field generated by the cell potential across perforations at this disruption would break the hydrogen bonds securing the gB fusion loops, leading to fusion of the plasma and viral membranes. This mechanism agrees with the high stability of the tall trimeric spike structure of gB and is consistent with the probable existence of a more compact initial conformation that would allow it to closely approach the plasma membrane. The release of the fusion domains by disruption of hydrogen bonds is shared with the endocytotic entry pathway where, for some cell types not punctured by gB, the virus is able to induce inward forces that cause endocytosis and the fusion loops are released by acidification. The puncture-fusion mechanism requires low critical strain or high tissue strain, matching primary tropism of neural processes at the vermillion border. In support of this mechanism, this paper proposes a functional superstructure of the antigens essential to entry and reviews its consistency with experimental evidence.
Collapse
Affiliation(s)
- Richard W. Clarke
- Chemistry Department, Cambridge University, Lensfield Road, Cambridge CB21EW, United Kingdom
| |
Collapse
|
46
|
Yang H, Li S, Li F, Wen R, Xiang J. Analysis on the expression and function of syndecan in the Pacific white shrimp Litopenaeus vannamei. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 51:278-286. [PMID: 25847874 DOI: 10.1016/j.dci.2015.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/31/2015] [Accepted: 03/31/2015] [Indexed: 06/04/2023]
Abstract
Syndecan is considered to be a multifunctional protein which functions as a cell surface receptor involved in cell adhesion, migration, cytoskeleton organization and differentiation. Previous bioinformatic analysis has revealed that syndecan in shrimp might interact with white spot syndrome virus (WSSV). In the present study, we experimentally studied the function of syndecan in shrimp immunity. The syndecan from Litopenaeus vannamei (LvSDC) was cloned and analyzed. The full-length cDNA of LvSDC was 1005 bp, consisting of 59 bp 5'-UTR, 253 bp 3'-UTR, and 693 bp open reading frame encoding 230 amino acids. LvSDC consisted of an extracellular domain (ED), a transmembrane domain (TM) and a cytoplasmic domain (CD). TM and CD shared high similarities with those of syndecan proteins from other species. LvSDC was ubiquitously expressed in all tested tissues, with the highest level in Oka. After WSSV challenge, the transcription level of LvSDC in Oka was apparently up-regulated. Recombinant LvSDC protein and its rabbit polyclonal antibody were prepared for detecting the location of LvSDC in hemocytes using immunocytochemistry approach. Data showed that LvSDC mainly located at the cell membrane and the cytoplasm of hemocytes. After silencing of LvSDC with siRNA, the WSSV copy numbers and mortality of shrimp after WSSV infection were both significantly decreased. These data provide useful information for understanding the immune mechanism of shrimp to WSSV infection.
Collapse
Affiliation(s)
- Hui Yang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shihao Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, 7 Nanhai Road, Qingdao 266071, China
| | - Fuhua Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, 7 Nanhai Road, Qingdao 266071, China.
| | - Rong Wen
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Jianhai Xiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
47
|
Heparanase is a host enzyme required for herpes simplex virus-1 release from cells. Nat Commun 2015; 6:6985. [PMID: 25912399 PMCID: PMC4413471 DOI: 10.1038/ncomms7985] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 03/23/2015] [Indexed: 12/27/2022] Open
Abstract
Herpesviruses exemplified by herpes simplex virus-1 (HSV-1) attach to cell surface heparan sulfate (HS) for entry into host cells. However, during a productive infection the HS moieties on parent cells can trap newly exiting viral progenies and inhibit their release. Here, we demonstrate that a HS-degrading enzyme of the host, heparanase (HPSE), is upregulated through NF-kB and translocated to the cell surface upon HSV-1 infection for the removal of HS to facilitate viral release. We also find a significant increase in HPSE release in vivo during infection of murine corneas and that knockdown of HPSE in vivo inhibits virus shedding. Overall, we propose that HPSE acts as a molecular switch for turning a virus-permissive “attachment mode” of host cells to a virus-deterring “detachment mode”. Since many human viruses use HS as an attachment receptor, the HPSE-HS interplay may delineate a common mechanism for virus release.
Collapse
|
48
|
Irvin SC, Herold BC. Molecular mechanisms linking high dose medroxyprogesterone with HIV-1 risk. PLoS One 2015; 10:e0121135. [PMID: 25798593 PMCID: PMC4370479 DOI: 10.1371/journal.pone.0121135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/11/2015] [Indexed: 11/24/2022] Open
Abstract
Background Epidemiological studies suggest that medroxyprogesterone acetate (MPA) may increase the risk of HIV-1. The current studies were designed to identify potential underlying biological mechanisms. Methods Human vaginal epithelial (VK2/E6E7), peripheral blood mononuclear (PBMC), and polarized endometrial (HEC-1-A) cells were treated with a range of concentrations of MPA (0.015-150 μg/ml) and the impact on gene expression, protein secretion, and HIV infection was evaluated. Results Treatment of VK2/E6E7 cells with high doses (>15μg/ml] of MPA significantly upregulated proinflammatory cytokines, which resulted in a significant increase in HIV p24 levels secreted by latently infected U1 cells following exposure to culture supernatants harvested from MPA compared to mock-treated cells. MPA also increased syndecan expression by VK2/E6E7 cells and cells treated with 15 μg/ml of MPA bound and transferred more HIV-1 to T cells compared to mock-treated cells. Moreover, MPA treatment of epithelial cells and PBMC significantly decreased cell proliferation resulting in disruption of the epithelial barrier and decreased cytokine responses to phytohaemagglutinin, respectively. Conclusion We identified several molecular mechanisms that could contribute to an association between DMPA and HIV including proinflammatory cytokine and chemokine responses that could activate the HIV promoter and recruit immune targets, increased expression of syndecans to facilitate the transfer of virus from epithelial to immune cells and decreased cell proliferation. The latter could impede the ability to maintain an effective epithelial barrier and adversely impact immune cell function. However, these responses were observed primarily following exposure to high (15-150 μg/ml) MPA concentrations. Clinical correlation is needed to determine whether the prolonged MPA exposure associated with contraception activates these mechanisms in vivo.
Collapse
Affiliation(s)
- Susan C. Irvin
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America
| | - Betsy C. Herold
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, United States of America
- * E-mail:
| |
Collapse
|
49
|
Tiwari V, Tarbutton MS, Shukla D. Diversity of heparan sulfate and HSV entry: basic understanding and treatment strategies. Molecules 2015; 20:2707-27. [PMID: 25665065 PMCID: PMC6272628 DOI: 10.3390/molecules20022707] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/02/2015] [Indexed: 12/30/2022] Open
Abstract
A modified form of heparan sulfate (HS) known as 3-O-sulfated heparan sulfate (3-OS HS) generates fusion receptor for herpes simplex virus (HSV) entry and spread. Primary cultures of corneal fibroblasts derived from human eye donors have shown the clinical significance of this receptor during HSV corneal infection. 3-OS HS- is a product of a rare enzymatic modification at C3 position of glucosamine residue which is catalyzed by 3-O-sulfotransferases (3-OSTs) enzymes. From humans to zebrafish, the 3-OST enzymes are highly conserved and widely expressed in cells and tissues. There are multiple forms of 3-OSTs each producing unique subset of sulfated HS making it chemically diverse and heterogeneous. HSV infection of cells or zebrafish can be used as a unique tool to understand the structural-functional activities of HS and 3-OS HS and likewise, the infection can be used as a functional assay to screen phage display libraries for identifying HS and 3-OS HS binding peptides or small molecule inhibitors. Using this approach over 200 unique 12-mer HS and 3-OS HS recognizing peptides were isolated and characterized against HSV corneal infection where 3-OS HS is known to be a key receptor. In this review we discuss emerging role of 3-OS HS based therapeutic strategies in preventing viral infection and tissue damage.
Collapse
Affiliation(s)
- Vaibhav Tiwari
- Department of Microbiology & Immunology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA.
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Morgan S Tarbutton
- Department of Microbiology & Immunology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA.
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA.
- Department of Microbiology & Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
50
|
Abstract
INTRODUCTION Almost half the global population is estimated to be at risk of contracting dengue infection. Of the 400 million infections estimated to occur annually, 4 million can be potentially life-threatening leading to vascular leakage and shock. The only treatment available to severe dengue patients is fluid replacement therapy and supportive care. A drug for treating dengue is an urgent need. AREAS COVERED This article endeavors to provide an overview of the experimental dengue drugs being developed around the world as reflected in the recent patent literature spanning the last few years (2010-2014). EXPERT OPINION Dengue drug development is essentially in its infancy and currently hobbled by multiple factors including a poor understanding of the molecular mechanism of severe disease and lack of reliable small animal model for preclinical drug evaluation. More intense R&D coupled to setting up product development partnerships to facilitate the efficient movement of a drug molecule from the laboratory to the clinic is needed to make antiviral therapy for dengue a reality in the coming future.
Collapse
Affiliation(s)
- Hemalatha Beesetti
- Birla Institute of Technology and Science Pilani, Department of Biological Sciences , Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal, Ranga Reddy District, Hyderabad-500078 , India +91 40 66303631 ; +91 40 66303998 ; ,
| | | | | |
Collapse
|