1
|
Waxman B, Salka K, Timilsina U, Umthong S, Shukla D, Stavrou S. Heparanase, a host gene that potently restricts retrovirus transcription. mBio 2025; 16:e0325224. [PMID: 39998209 PMCID: PMC11980367 DOI: 10.1128/mbio.03252-24] [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: 10/18/2024] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Heparanase (HPSE) is a heterodimeric β-D-glucuronidase that is critical in mammalian cells for the enzymatic cleavage of membrane-associated heparan sulfate moieties. Apart from its enzymatic function, HPSE has important non-enzymatic functions, which include transcriptional regulation, chromatin modification, and modulation of various signaling pathways. Interestingly, while HPSE is an interferon-stimulated gene, past reports have shown that it has proviral properties for many different viruses, including herpes simplex virus 1, as it assists virus release from infected cells. However, as of yet, no antiviral functions associated with HPSE have been described. Here, we show that HPSE utilizes a hitherto unknown mechanism to restrict retroviruses by targeting the step of proviral transcription. Moreover, we demonstrate that HPSE blocks transcription initiation by targeting the SP1 transcription factor. Finally, we illustrate that the antiretroviral effect of HPSE is independent of its enzymatic activity. This report describes a novel antiviral mechanism utilized by HPSE to inhibit retrovirus infection.IMPORTANCEHeparanase (HPSE) has emerged as an important factor that has proviral functions for a number of viruses, including herpes simplex virus and hepatitis C virus, by assisting in virus egress. However, HPSE is an interferon-stimulated gene and, thus, is a part of the host antiviral defense. Nothing is known about the antiviral functions of HPSE. Here, we examine in depth the role of HPSE during retrovirus infection using two retroviruses, human immunodeficiency virus type 1 (HIV-1) and murine leukemia virus. In this report, we show that mouse, but not human, HPSE blocks retrovirus infection by targeting provirus transcription. HPSE sequesters the SP1 transcription factor away from the proviral promoter, thereby inhibiting transcription initiation. In conclusion, our findings identify a novel antiviral function of HPSE and its potential role as an inhibitor of zoonotic transmission of retroviruses.
Collapse
Affiliation(s)
- Brandon Waxman
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, USA
| | - Kyle Salka
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, USA
| | - Uddhav Timilsina
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, USA
| | - Supawadee Umthong
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, USA
| | - Spyridon Stavrou
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, USA
| |
Collapse
|
2
|
Borase H, Patil CD, Valyi-Nagy T, Shukla D. HPSE-mediated proinflammatory signaling contributes to neurobehavioral deficits following intranasal HSV-1 infection. mBio 2025; 16:e0376524. [PMID: 40013778 PMCID: PMC11980599 DOI: 10.1128/mbio.03765-24] [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: 12/10/2024] [Accepted: 01/16/2025] [Indexed: 02/28/2025] Open
Abstract
Herpes simplex virus-1 (HSV-1) is a neurotropic virus that can infect the brain, and an uncontrolled infection can lead to a range of diseases, including chronic nerve pain, encephalitis, and neurobehavioral abnormalities. These outcomes are often severe and have lasting consequences, highlighting the need to identify host factors that contribute to disease severity. In this study, we report that intranasal HSV-1 infection in murine model, which promotes viral dissemination into the brain, implicates the host protein heparanase (HPSE) as a key mediator of neuroinflammation. Specifically, we observed that the HPSE activity during HSV-1 infection in naïve animals promotes the upregulation of proinflammatory cytokines, enhances microglial activity in the brain, and contributes to cognitive impairment, anxiety, and motor coordination deficits. Such effects are significantly less detectable in heparanase deficient (Hpse-/-) mice. Additionally, we found that moderate activation of toll-like receptors (TLRs), particularly in Hpse+/+ mice, may contribute to the activation of the inflammasome pathway. This, in turn, leads to the activation of caspase-1 (Casp1) and caspase-3 (Casp3), which may play a role in nerve function loss. Our findings position HPSE as a potential therapeutic target for mitigating virus-induced neuroinflammation and neurobehavioral defects. IMPORTANCE Herpes simplex virus-1 (HSV-1) infection in the brain can lead to severe and often permanent neurological consequences. Host factors influence disease outcomes in response to infection, and understanding these factors is crucial for developing effective therapies. This study identifies the host protein HPSE as a key mediator of neuroinflammation in response to HSV-1 infection. We demonstrate that the HPSE activity drives proinflammatory cytokine expression and microglial activation and promotes a signaling cascade involving toll-like receptors and caspase activation, potentially intensifying neuroinflammatory responses. These findings implicate HPSE as an important player in HSV-1 pathogenesis in the central nervous system and suggest that targeting HPSE could provide a novel therapeutic strategy to mitigate virus-induced neuroinflammation and neurobehavioral disturbance.
Collapse
Affiliation(s)
- Hemant Borase
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, USA
| | - Chandrashekhar D. Patil
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, USA
| | - Tibor Valyi-Nagy
- Department of Pathology, Neuropathology Service, University of Illinois Chicago, Chicago, Illinois, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Wang J, Rabiee B, Patel C, Jafri M, Hussain H, Chaudhry A, Chaudhry I, Kamoun L, Chaudhry I, Oh L, Bobat FI, Shukla D, Farooq AV. Herpesvirus Infections of the Corneal Endothelium. Microorganisms 2025; 13:778. [PMID: 40284615 PMCID: PMC12029634 DOI: 10.3390/microorganisms13040778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Corneal endotheliitis is an inflammatory process, most commonly of viral etiology, that manifests clinically with features including corneal edema, keratic precipitates, and a mild anterior chamber reaction. Several studies have implicated human herpesviruses from the Herpesviridae family as primary causes of corneal endotheliitis, including cytomegalovirus (CMV), varicella zoster virus (VZV), and herpes simplex viruses 1 and 2 (HSV-1 and HSV-2). This review critically evaluates the present literature surrounding herpesvirus infections of the corneal endothelium.
Collapse
Affiliation(s)
- Jessie Wang
- Duke Eye Center, Duke University, Durham, NC 27705, USA;
| | - Behnam Rabiee
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Chandani Patel
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Mansab Jafri
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Hamad Hussain
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Aaila Chaudhry
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Imtiaz Chaudhry
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Layla Kamoun
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Iftikhar Chaudhry
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Lewis Oh
- Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Fatima I. Bobat
- Department of Ophthalmology and Visual Sciences, The University of Illinois Chicago, Chicago, IL 60607, USA; (F.I.B.); (D.S.)
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, The University of Illinois Chicago, Chicago, IL 60607, USA; (F.I.B.); (D.S.)
| | - Asim V. Farooq
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
4
|
Sharma P, Naqvi RA, Borase H, Kapoor D, Valverde A, Capistrano K, Yadavalli T, Naqvi AR, Shukla D. Global MicroRNA Profiling of HSV-1 Infected Cornea Identifies miR-329 as a Novel Regulator of Virus Infection. Invest Ophthalmol Vis Sci 2025; 66:61. [PMID: 39992671 PMCID: PMC11878248 DOI: 10.1167/iovs.66.2.61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Purpose Although the mechanisms underlying herpes simplex virus type-1 (HSV-1) ocular infection have been extensively studied, the role of host microRNAs (miRNAs) in the pathobiology of herpetic keratitis (HK) is not well understood. The aim of this study was to identify endogenous miRNA regulators involved in the progression of HSV-1 ocular infection. Methods C57BL/6 mice were infected with HSV-1 strain McKrae following epithelial debridement, and corneal miRNA profiles were analyzed at various time points using miRNA sequencing (miRNA-seq). The miRNA expression was measured at 2, 4, 6, and 10 days post-infection. Ingenuity Pathway Analysis (IPA) was used to identify immune pathways potentially targeted by differentially expressed miRNAs. The role of selected miRNAs in viral entry and replication was assessed by overexpression in murine embryonic fibroblasts (MEFs) and human corneal epithelial cells (HCEs). Results A total of 32 miRNAs at 2 days post-infection, 21 miRNAs at 4 days post-infection, 140 miRNAs at 6 days post-infection, and 27 miRNAs at 10 days post-infection showed significant changes in expression. IPA revealed that differentially expressed miRNAs targeted several immune pathways, including TLR and interferon signaling. Notably, mmu-miR-184-3p and mmu-let-7d-5p were upregulated, whereas mmu-miR-329-3p was down-regulated during infection. Functional assays demonstrated that overexpression of miR-329, but not miR-184-3p or miR-let-7d-5p, increased HSV-1 viral entry and replication in a dose-dependent manner. In contrast, miR-329 inhibition reversed these effects, suggesting its role as a pro-viral miRNA. Increased plaque formation and viral gB expression further confirmed miR-329's pro-viral role. Conclusions Our findings suggest that miR-329 functions as a pro-viral miRNA by disrupting TLR9 signaling, thus facilitating HSV-1 replication. Inhibition of miR-329 enhances TLR9-mediated antiviral responses, highlighting the potential of targeting host miRNAs as a novel therapeutic strategy for managing viral keratitis.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/biosynthesis
- Animals
- Herpesvirus 1, Human/physiology
- Herpesvirus 1, Human/genetics
- Keratitis, Herpetic/genetics
- Keratitis, Herpetic/virology
- Keratitis, Herpetic/metabolism
- Mice, Inbred C57BL
- Mice
- Humans
- Virus Replication
- Gene Expression Profiling
- Epithelium, Corneal/virology
- Epithelium, Corneal/metabolism
- Disease Models, Animal
- Gene Expression Regulation/physiology
- Cornea/virology
- Cornea/metabolism
- Cells, Cultured
Collapse
Affiliation(s)
- Pankaj Sharma
- Department of Ophthalmology, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Raza Ali Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Hemant Borase
- Department of Ophthalmology, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Divya Kapoor
- Department of Ophthalmology, University of Illinois - Chicago, Chicago, Illinois, United States
- Department of Microbiology and Immunology, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Araceli Valverde
- Department of Periodontics, College of Dentistry, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Kristelle Capistrano
- Department of Periodontics, College of Dentistry, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Tejabhiram Yadavalli
- Department of Ophthalmology, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Afsar R. Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois - Chicago, Chicago, Illinois, United States
| | - Deepak Shukla
- Department of Ophthalmology, University of Illinois - Chicago, Chicago, Illinois, United States
- Department of Microbiology and Immunology, University of Illinois - Chicago, Chicago, Illinois, United States
| |
Collapse
|
5
|
Hopkins J, Volety I, Qatanani F, Shukla D. Heparanase 2 Modulation Inhibits HSV-2 Replication by Regulating Heparan Sulfate. Viruses 2024; 16:1832. [PMID: 39772142 PMCID: PMC11680312 DOI: 10.3390/v16121832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/17/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
The host enzyme heparanase (HPSE) facilitates the release of herpes simplex virus type 2 (HSV-2) from target cells by cleaving the viral attachment receptor heparan sulfate (HS) from infected cell surfaces. HPSE 2, an isoform of HPSE, binds to but does not possess the enzymatic activity needed to cleave cell surface HS. Our study demonstrates that HSV-2 infection significantly elevates HPSE 2 protein levels, impacting two distinct stages of viral replication. We show that higher HPSE 2 negatively affects HSV-2 replication which may be through the regulation of cell surface HS. By acting as a competitive inhibitor of HPSE, HPSE 2 may be interfering with HPSE's interactions with HS. We demonstrate that the enhanced expression of HPSE 2, either via viral infection or plasmid transfection, reduces HPSE's ability to cleave HS, thereby hindering viral egress. Conversely, low HPSE 2 levels achieved through siRNA transfection allow HPSE to cleave more HS, reducing viral entry. Altogether, we propose a hypothetical model in which the modulation of HPSE 2 impedes HSV-2 replication by regulating HS availability on the cell surface. This dual role of HPSE 2 in viral replication and potential tumor suppression underscores its significance in cellular processes and viral pathogenesis.
Collapse
Affiliation(s)
- James Hopkins
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (J.H.); (I.V.); (F.Q.)
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ipsita Volety
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (J.H.); (I.V.); (F.Q.)
- Department of Pathology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Farreh Qatanani
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (J.H.); (I.V.); (F.Q.)
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (J.H.); (I.V.); (F.Q.)
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
6
|
Manouchehri JM, Datta J, Marcho LM, Reardon JJ, Stover D, Wesolowski R, Borate U, Cheng TYD, Schnell PM, Ramaswamy B, Sizemore GM, Rubinstein MP, Cherian MA. The role of heparan sulfate in enhancing the chemotherapeutic response in triple-negative breast cancer. Breast Cancer Res 2024; 26:153. [PMID: 39506780 PMCID: PMC11539583 DOI: 10.1186/s13058-024-01906-6] [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: 04/17/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Breast cancer, one of the most common forms of cancer, is associated with the highest cancer-related mortality among women worldwide. In comparison to other types of breast cancer, patients diagnosed with the triple-negative breast cancer (TNBC) subtype have the worst outcome because current therapies do not produce long-lasting responses. Hence, innovative therapies that produce persisting responses are a critical need. We previously discovered that hyperactivating purinergic receptors (P2RXs) by increasing extracellular adenosine triphosphate (eATP) concentrations enhances TNBC cell lines' response to chemotherapy. Heparan sulfate inhibits multiple extracellular ATPases, so it is a molecule of interest in this regard. In turn, heparanase degrades polysulfated polysaccharide heparan sulfate. Importantly, previous work suggests that breast cancer and other cancers express heparanase at high levels. Hence, as heparan sulfate can inhibit extracellular ATPases to facilitate eATP accumulation, it may intensify responses to chemotherapy. We postulated that heparanase inhibitors would exacerbate chemotherapy-induced decreases in TNBC cell viability by increasing heparan sulfate in the cellular microenvironment and hence, augmenting eATP. METHODS We treated TNBC cell lines MDA-MB 231, Hs 578t, and MDA-MB 468 and non-tumorigenic immortal mammary epithelial MCF-10A cells with paclitaxel (cytotoxic chemotherapeutic) with or without the heparanase inhibitor OGT 2115 and/or supplemental heparan sulfate. We evaluated cell viability and the release of eATP. Also, we compared the expression of heparanase protein in cell lines and tissues by immunoblot and immunohistochemistry, respectively. In addition, we examined breast-cancer-initiating cell populations using tumorsphere formation efficiency assays on treated cells. RESULTS We found that combining heparanase inhibitor OGT 2115 with chemotherapy decreased TNBC cell viability and tumorsphere formation through increases in eATP and activation of purinergic receptors as compared to TNBC cells treated with single-agent paclitaxel. CONCLUSION Our data shows that by preventing heparan sulfate breakdown, heparanase inhibitors make TNBC cells more susceptible to chemotherapy by enhancing eATP concentrations.
Collapse
Affiliation(s)
- Jasmine M Manouchehri
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jharna Datta
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Lynn M Marcho
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jesse J Reardon
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Daniel Stover
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Uma Borate
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Ting-Yuan David Cheng
- Division of Cancer Prevention and Control, Department of Internal Medicine, The Ohio State University, Suite 525, 1590 North High St., Columbus, OH, 43201, USA
| | - Patrick M Schnell
- Division of Biostatistics, The Ohio State University College of Public Health, 1841 Neil Ave., Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Gina M Sizemore
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mark P Rubinstein
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mathew A Cherian
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA.
- Division of Medical Oncology, 460 W 12th Ave., 888 BRT, Columbus, OH, 43210, USA.
| |
Collapse
|
7
|
De Arrigunaga S, Wall S, Theotoka D, Friehmann A, Camacho M, Dubovy S, Galor A, Karp CL. Chronic inflammation as a proposed risk factor for ocular surface squamous neoplasia. Ocul Surf 2024; 33:1-10. [PMID: 38432640 DOI: 10.1016/j.jtos.2024.02.006] [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: 04/26/2023] [Revised: 12/19/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE Chronic inflammation is a predisposing factor for metaplastic changes and ultimately dysplasia. We describe cases of OSSN occurring in the setting of chronic ocular surface inflammation. METHODS Sixteen eyes from 14 individuals were included from one ocular oncology clinic between 2010 and 2023. Patients presented with ocular surface squamous neoplasia (OSSN) in the setting of chronic inflammation. The diagnosis of OSSN was made using anterior segment high-resolution optical coherence tomography (HR-OCT) and confirmed by histopathological analysis in all cases. RESULTS Median age on presentation was 61 [IQR 47.5-69.2] years. Eleven (86%) individuals were male and five (36%) identified as White Hispanic. Ten eyes were referred with ocular surface diagnoses including pannus (n = 4), scarring (n = 3), pterygium (n = 2), and herpetic keratitis (n = 1). Only six eyes were referred as possible neoplasia. All individuals had a history of ocular surface inflammation. The most common inflammatory conditions were ocular rosacea (seven individuals) and atopic keratoconjunctivitis (AKC) (five individuals). Two individuals were found to have bilateral OSSN, one in the setting of ocular rosacea and the other in the setting of AKC. All 16 eyes from 14 individuals were suspected to have OSSN based on HR-OCT findings which guided the location of the incisional biopsies that subsequently confirmed histopathological diagnosis in all cases. CONCLUSION OSSN may arise in the setting of chronic inflammation on the ocular surface. Identification of the tumor can be challenging in these cases, and HR-OCT can be a key diagnostic tool in detecting OSSN.
Collapse
Affiliation(s)
- Sofia De Arrigunaga
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA
| | - Sarah Wall
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology and Visual Science, Yale School of Medicine, 40 Temple Street, New Haven, CT, 06510, USA
| | - Despoina Theotoka
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology and Visual Science, Yale School of Medicine, 40 Temple Street, New Haven, CT, 06510, USA
| | - Asaf Friehmann
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology, Meir Medical Center, Kfar Saba, Israel; Sackler faculty of Medicine, Tel Aviv University, Israel
| | - Matthew Camacho
- Florida Lions Ocular Pathology Laboratory, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sander Dubovy
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Florida Lions Ocular Pathology Laboratory, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anat Galor
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA; Department of Ophthalmology, Miami Veterans Administration Medical Center, Miami, FL, USA
| | - Carol L Karp
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 900 NW 17th St, Miami, FL, 33136, USA.
| |
Collapse
|
8
|
Kapoor D, Sharma P, Shukla D. Emerging drugs for the treatment of herpetic keratitis. Expert Opin Emerg Drugs 2024; 29:113-126. [PMID: 38603466 DOI: 10.1080/14728214.2024.2339899] [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: 02/07/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Herpes simplex keratitis stands as a prominent factor contributing to infectious blindness among developed nations. On a global scale, over 60% of the population tests positive for herpes simplex virus type-1 (HSV-1). Despite these statistics, there is currently no vaccine available for the virus. Moreover, the conventional nucleoside drugs prescribed to patients are proving ineffective in addressing issues related to drug resistance, recurrence, latency, and the escalating risk of vision loss. Hence, it is imperative to continually explore all potential avenues to restrict the virus. This review article centers on the present treatment methods for HSV-1 keratitis (HSK), highlighting the ongoing clinical trials. It delves into the emerging drugs, their mode-of-action and future therapeutics. AREAS COVERED The review focuses on the significance of a variety of small molecules targeting HSV-1 lifecycle at multiple steps. Peer-reviewed articles and abstracts were searched in MEDLINE, PubMed, Embase, and clinical trial websites. EXPERT OPINION The exploration of small molecules that target specific pathways within the herpes lifecycle holds the potential for substantial impact on the antiviral pharmaceutical market. Simultaneously, the pursuit of disease-specific biomarkers has the capacity to usher in a transformative era in diagnostics within the field.
Collapse
Affiliation(s)
- Divya Kapoor
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, USA
| | - Pankaj Sharma
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, USA
| |
Collapse
|
9
|
Katz M, Diskin R. The underlying mechanisms of arenaviral entry through matriglycan. Front Mol Biosci 2024; 11:1371551. [PMID: 38516183 PMCID: PMC10955480 DOI: 10.3389/fmolb.2024.1371551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Matriglycan, a recently characterized linear polysaccharide, is composed of alternating xylose and glucuronic acid subunits bound to the ubiquitously expressed protein α-dystroglycan (α-DG). Pathogenic arenaviruses, like the Lassa virus (LASV), hijack this long linear polysaccharide to gain cellular entry. Until recently, it was unclear through what mechanisms LASV engages its matriglycan receptor to initiate infection. Additionally, how matriglycan is synthesized onto α-DG by the Golgi-resident glycosyltransferase LARGE1 remained enigmatic. Recent structural data for LARGE1 and for the LASV spike complex informs us about the synthesis of matriglycan as well as its usage as an entry receptor by arenaviruses. In this review, we discuss structural insights into the system of matriglycan generation and eventual recognition by pathogenic viruses. We also highlight the unique usage of matriglycan as a high-affinity host receptor compared with other polysaccharides that decorate cells.
Collapse
Affiliation(s)
| | - Ron Diskin
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
10
|
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
|
11
|
Rabinowitz ZM, Wang Z, Liu J, Zhang Y, Ybargollin AJ, Saketkhou M, Cui L. A Fluorogenic Green Merocyanine-Based Probe to Detect Heparanase-1 Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.25.581963. [PMID: 38464176 PMCID: PMC10925095 DOI: 10.1101/2024.02.25.581963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Heparanase-1 (HPSE-1), an endo-β-D-glucuronidase, is an extracellular matrix (ECM) remodeling enzyme that degrades heparan sulfate (HS) chains of heparan sulfate proteoglycans (HSPGs). HPSE-1 functions to remodel the ECM and thereby disseminate cells, liberate HS-bound bioactive molecules, and release biologically active HS fragments. Being the only known enzyme for the cleavage of HS, HPSE-1 regulates a number of fundamental cellular processes including cell migration, cytokine regulation, angiogenesis, and wound healing. Overexpression of HPSE-1 has been discovered in most cancers, inflammatory diseases, viral infections, among others. As an emerging therapeutic target, the biological role of HPSE-1 remains to be explored but is hampered by a lack of research tools. To expand the chemical tool-kit of fluorogenic probes to interrogate HPSE-1 activity, we design and synthesized a fluorogenic green disaccharide-based HPSE-1 probe using our design strategy of tuning the electronic effect of the aryl aglycon. The novel probe exhibits a highly sensitive 278-fold fluorescence turn-on response in the presence of recombinant human HPSE-1, while emitting green light at 560 nm, enabling the fluorescence imaging of HPSE-1 activity in cells.
Collapse
|
12
|
Naqvi RA, Valverde A, Yadavalli T, Bobat FI, Capistrano KJ, Shukla D, Naqvi AR. Viral MicroRNAs in Herpes Simplex Virus 1 Pathobiology. Curr Pharm Des 2024; 30:649-665. [PMID: 38347772 DOI: 10.2174/0113816128286469240129100313] [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: 10/10/2023] [Accepted: 01/17/2024] [Indexed: 06/01/2024]
Abstract
Simplexvirus humanalpha1 (Herpes simplex virus type 1 [HSV-1]) infects millions of people globally, manifesting as vesiculo-ulcerative lesions of the oral or genital mucosa. After primary infection, the virus establishes latency in the peripheral neurons and reactivates sporadically in response to various environmental and genetic factors. A unique feature of herpesviruses is their ability to encode tiny noncoding RNAs called microRNA (miRNAs). Simplexvirus humanalpha1 encodes eighteen miRNA precursors that generate twentyseven different mature miRNA sequences. Unique Simplexvirus humanalpha1 miRNAs repertoire is expressed in lytic and latent stages and exhibits expressional disparity in various cell types and model systems, suggesting their key pathological functions. This review will focus on elucidating the mechanisms underlying the regulation of host-virus interaction by HSV-1 encoded viral miRNAs. Numerous studies have demonstrated sequence- specific targeting of both viral and host transcripts by Simplexvirus humanalpha1 miRNAs. While these noncoding RNAs predominantly target viral genes involved in viral life cycle switch, they regulate host genes involved in antiviral immunity, thereby facilitating viral evasion and lifelong viral persistence inside the host. Expression of Simplexvirus humanalpha1 miRNAs has been associated with disease progression and resolution. Systemic circulation and stability of viral miRNAs compared to viral mRNAs can be harnessed to utilize their potential as diagnostic and prognostic markers. Moreover, functional inhibition of these enigmatic molecules may allow us to devise strategies that have therapeutic significance to contain Simplexvirus humanalpha1 infection.
Collapse
Affiliation(s)
- Raza Ali Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Araceli Valverde
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, Medical Center, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Fatima Ismail Bobat
- Department of Ophthalmology and Visual Sciences, Medical Center, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Kristelle J Capistrano
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, Medical Center, University of Illinois Chicago, Chicago, Illinois 60607, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Afsar R Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, Illinois 60607, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
13
|
Yadavalli T, Patil C, Sharma P, Volety I, Borase H, Kapoor D, Shukla D. Unique Attributes of Guinea Pigs as New Models to Study Ocular Herpes Pathophysiology and Recurrence. Invest Ophthalmol Vis Sci 2023; 64:41. [PMID: 38015175 PMCID: PMC10691389 DOI: 10.1167/iovs.64.14.41] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/29/2023] Open
Abstract
Purpose The objective of this study was to explore the ocular and systemic outcomes of herpes simplex virus type 1 (HSV-1) infection in guinea pigs, to monitor the spontaneous reactivation of the virus, and to assess the effectiveness of various treatments, drawing comparisons to conventional rabbit models. Methods Guinea pigs and rabbits were infected in the right corneas with differing doses and strains of HSV-1. Observations were made over a 71-day period, focusing on comparing ocular lesions, viral shedding patterns, and weight loss between the two animal models. Postinfection, the effectiveness of trifluridine ophthalmic drops, oral acyclovir, and valacyclovir was evaluated. The confirmation of viral infection was done through virus titer assay, fluorescein staining, and corneal imaging. Results Guinea pigs and rabbits manifested symptoms akin to human herpes stromal keratitis (HSK) when exposed to varying titers of viral suspension. Regardless of the initial viral load, all guinea pig groups demonstrated comparable ocular pathology, witnessing conditions like blepharitis and conjunctivitis within 3 days, progressing to severe conditions, including total corneal opacification and necrotizing keratitis. Tear film collection revealed nonsignificant differences in viral plaques between all groups. Notably, guinea pigs in the low-infection group experienced the most weight loss, although without significant differences. The replication of the same experiment on rabbits yielded consistent results in disease pathology across different groups, with occurrences of blepharitis and conjunctivitis. Interestingly, after initial resolution, guinea pigs presented a more frequent and broadly observed increase in disease score and corneal opacity, a phenomenon rarely seen in rabbits within the same timeframe. The effectiveness of 1% trifluridine was observed in mitigating ocular HSV-1 disease in both species, whereas oral acyclovir and valacyclovir were found to be detrimental and ineffective in guinea pigs but not in rabbits. Conclusions This study demonstrates the potential suitability of guinea pigs as new models for ocular HSV-1 investigations, filling a critical preclinical void of models capable of showcasing spontaneous HSV reactivation in the eye. The observed similarities and differences in the reactions of guinea pigs and rabbits to HSV-1 infection and treatments provide crucial insights, laying the foundation for future studies on ocular HSV pathogenesis, latency, and improved treatment options.
Collapse
Affiliation(s)
- Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, United States
| | - Chandrashekhar Patil
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, United States
| | - Pankaj Sharma
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, United States
| | - Ipsita Volety
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, United States
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, United States
| | - Hemant Borase
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, United States
| | - Divya Kapoor
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, United States
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Chicago, Illinois, United States
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States
| |
Collapse
|
14
|
Chopra P, Yadavalli T, Palmieri F, Jongkees SAK, Unione L, Shukla D, Boons GJ. Synthetic Heparanase Inhibitors Can Prevent Herpes Simplex Viral Spread. Angew Chem Int Ed Engl 2023; 62:e202309838. [PMID: 37555536 DOI: 10.1002/anie.202309838] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/10/2023]
Abstract
Herpes simplex virus (HSV-1) employs heparan sulfate (HS) as receptor for cell attachment and entry. During late-stage infection, the virus induces the upregulation of human heparanase (Hpse) to remove cell surface HS allowing viral spread. We hypothesized that inhibition of Hpse will prevent viral release thereby representing a new therapeutic strategy for HSV-1. A range of HS-oligosaccharides was prepared to examine the importance of chain length and 2-O-sulfation of iduronic moieties for Hpse inhibition. It was found that hexa- and octasaccharides potently inhibited the enzyme and that 2-O-sulfation of iduronic acid is tolerated. Computational studies provided a rationale for the observed structure-activity relationship. Treatment of human corneal epithelial cells (HCEs) infected with HSV-1 with the hexa- and octasaccharide blocked viral induced shedding of HS which significantly reduced spread of virions. The compounds also inhibited migration and proliferation of immortalized HCEs thereby providing additional therapeutic properties.
Collapse
Affiliation(s)
- Pradeep Chopra
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Francesco Palmieri
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Seino A K Jongkees
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Luca Unione
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Current address: CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, 48160, Derio, Bizkaia, Spain
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Bijvoet Center for Biomolecular Research, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
15
|
Manouchehri JM, Marcho L, Cherian MA. The role of heparan sulfate in enhancing the chemotherapeutic response in triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556819. [PMID: 37745355 PMCID: PMC10515779 DOI: 10.1101/2023.09.08.556819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Background Among women worldwide, breast cancer has the highest incidence and is the leading cause of cancer-related death. Patients with the triple-negative breast cancer (TNBC) subtype have an inferior prognosis in comparison to other breast cancers because current therapies do not facilitate long-lasting responses. Thus, there is a demand for more innovative therapies that induce durable responses.In our previous research, we discovered that augmenting the concentration of extracellular ATP (eATP) greatly enhances the chemotherapeutic response of TNBC cell lines by activating purinergic receptors (P2RXs), leading to cell death through the induction of non-selective membrane permeability. However, eATP levels are limited by several classes of extracellular ATPases. One endogenous molecule of interest that can inhibit multiple classes of extracellular ATPases is heparan sulfate. Polysulfated polysaccharide heparan sulfate itself is degraded by heparanase, an enzyme that is known to be highly expressed in various cancers, including breast cancer. Heparan sulfate has previously been shown to regulate several cancer-related processes such as fibroblast growth factor signaling, neoangiogenesis by sequestering vascular endothelial growth factors in the extracellular matrix, hedgehog signaling and cell adhesion. In this project, we identified an additional mechanism for a tumor suppressor role of heparan sulfate: inhibition of extracellular ATPases, leading to augmented levels of eATP.Several heparanase inhibitors have been previously identified, including OGT 2115, suramin, PI-88, and PG 545. We hypothesized that heparanase inhibitors would augment eATP concentrations in TNBC by increasing heparan sulfate in the tumor microenvironment, resulting in enhanced cell death in response to chemotherapy. Methods We treated TNBC cell lines MDA-MB 231, Hs 578t, and MDA-MB 468 and non-tumorigenic immortal mammary epithelial MCF-10A cells with increasing concentrations of the chemotherapeutic agent paclitaxel in the presence of heparan sulfate and/or the heparanase inhibitor OGT 2115 while analyzing eATP release and cell viability. Moreover, to verify that the effects of OGT 2115 are mediated through eATP, we applied specific antagonists to the purinergic receptors P2RX4 and P2RX7. In addition, the protein expression of heparanase was compared in the cell lines by Western blot analysis. We also evaluated the consequences of this therapeutic strategy on the breast cancer-initiating cell population in the treated cells using flow cytometry and tumorsphere formation efficiency assays. Results Heparanase was found to be highly expressed in immortal mammary epithelial cells in comparison to TNBC cell lines. The heparanase inhibitor OGT 2115 augmented chemotherapy-induced TNBC cell death and eATP release. Conclusion These results demonstrate that inhibiting the degradation of heparan sulfate in the tumor microenvironment augments the susceptibility of TNBC cell lines to chemotherapy by increasing extracellular ATP concentrations. This strategy could potentially be applied to induce more enhanced and enduring responses in TNBC patients.
Collapse
|
16
|
Suryawanshi RK, Patil CD, Agelidis A, Koganti R, Yadavalli T, Ames JM, Borase H, Shukla D. Pathophysiology of reinfection by exogenous HSV-1 is driven by heparanase dysfunction. SCIENCE ADVANCES 2023; 9:eadf3977. [PMID: 37115924 PMCID: PMC10146881 DOI: 10.1126/sciadv.adf3977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/23/2023] [Indexed: 05/03/2023]
Abstract
Limited knowledge exists on exogenous DNA virus reinfections. Herpes simplex virus-1 (HSV-1), a prototype DNA virus, causes multiple human diseases including vision-threatening eye infections. While reinfection with an exogenous HSV-1 strain is considered plausible, little is known about the underlying mechanisms governing its pathophysiology in a host. Heparanase (HPSE), a host endoglycosidase, when up-regulated by HSV-1 infection dictates local inflammatory response by destabilizing tissue architecture. Here, we demonstrate that HSV-1 reinfection in mice causes notable pathophysiology in wild-type controls compared to the animals lacking HPSE. The endoglycosidase promotes infected cell survival and supports a pro-disease environment. In contrast, lack of HPSE strengthens intrinsic immunity by promoting cytokine expression, inducing necroptosis of infected cells, and decreasing leukocyte infiltration into the cornea. Collectively, we report that immunity from a recent prior infection fails to abolish disease manifestation during HSV-1 reinfection unless HPSE is rendered inactive.
Collapse
Affiliation(s)
- Rahul K. Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Chandrashekhar D. Patil
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Alex Agelidis
- 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
| | - Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Joshua M. Ames
- 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
| | - Hemant Borase
- Department of Ophthalmology and Visual Sciences, 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
|
17
|
Heparanase Modulates Chromatin Accessibility. Cells 2023; 12:cells12060891. [PMID: 36980232 PMCID: PMC10047235 DOI: 10.3390/cells12060891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/04/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Heparanase is the sole endoglucuronidase that degrades heparan sulfate in the cell surface and extracellular matrix (ECM). Several studies have reported the localization of heparanase in the cell nucleus, but the functional role of the nuclear enzyme is still obscure. Subjecting mouse embryonic fibroblasts (MEFs) derived from heparanase knockout (Hpse-KO) mice and applying transposase-accessible chromatin with sequencing (ATAC-seq), we revealed that heparanase is involved in the regulation of chromatin accessibility. Integrating with genome-wide analysis of chromatin states revealed an overall low activity in the enhancer and promoter regions of Hpse-KO MEFs compared with wild-type (WT) MEFs. Western blot analysis of MEFs and tissues derived from Hpse-KO vs. WT mice confirmed reduced expression of H3K27ac (acetylated lysine at N-terminal position 27 of the histone H3 protein). Our results offer a mechanistic explanation for the well-documented attenuation of inflammatory responses and tumor growth in Hpse-KO mice.
Collapse
|
18
|
Kapoor D, Shukla D. Neutrophil Extracellular Traps and Their Possible Implications in Ocular Herpes Infection. Pathogens 2023; 12:209. [PMID: 36839481 PMCID: PMC9958879 DOI: 10.3390/pathogens12020209] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are net-like structures released from neutrophils. NETs predominantly contain cell-free deoxyribonucleic acid (DNA) decorated with histones and neutrophil granule proteins. Numerous extrinsic and intrinsic stimuli can induce the formation of NETs such as pathogens, cytokines, immune complexes, microcrystals, antibodies, and other physiological stimuli. The mechanism of NETosis induction can either be ROS-dependent or independent based on the catalase producing activity of the pathogen. NADPH is the source of ROS production, which in turn depends on the upregulation of Ca2+ production in the cytoplasm. ROS-independent induction of NETosis is regulated through toll-like receptors (TLRs). Besides capturing and eliminating pathogens, NETs also aggravate the inflammatory response and thus act as a double-edged sword. Currently, there are growing reports of NETosis induction during bacterial and fungal ocular infections leading to different pathologies, but there is no direct report suggesting its role during herpes simplex virus (HSV) infection. There are innumerable independent reports showing that the major effectors of NETosis are also directly affected by HSV infection, and thus, there is a strong possibility that HSV interacts with these facilitators that can either result in virally mediated modulation of NETosis or NETosis-mediated suppression of ocular HSV infection. This review focuses on the mechanism of NETs formation during different ocular pathologies, with its prime focus on highlighting their potential implications during HSV ocular infections and acting as prospective targets for the treatment of ocular diseases.
Collapse
Affiliation(s)
- Divya Kapoor
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1905 W. Taylor St., Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1905 W. Taylor St., Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
| |
Collapse
|
19
|
Al-Kuraishy HM, Al-Gareeb AI, Hetta HF, Alexiou A, Papadakis M, Batiha GES. Heparanase is the possible link between monkeypox and Covid-19: robust candidature in the mystic and present perspective. AMB Express 2023; 13:13. [PMID: 36705773 PMCID: PMC9880376 DOI: 10.1186/s13568-023-01517-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Heparanase (HPSE) is an endoglycosidase cleaves heparan sulfate (HS) and this contributes to the degradation and remodeling of the extracellular matrix. HS cleaved by HPSE induces activation of autophagy and formation of autophagosommes which facilitate binding of HPSE to the HS and subsequent release of growth factors. The interaction between HPSE and HS triggers releases of chemokines and cytokines which affect inflammatory response and cell signaling pathways with development of hyperinflammation, cytokine storm (CS) and coagulopathy. HPSE expression is induced by both SARS-CoV-2 and monkeypox virus (MPXV) leading to induction release of pro-inflammatory cytokines, endothelial dysfunction and thrombotic events. Co-infection of MPX with SARS-CoV-2 may occur as we facing many outbreaks of MPX cases during Covid-19 pandemic. Therefore, targeting of HPSE by specific inhibitors may reduce the risk of complications in both SARS-CoV-2 and MPXV infections. Taken together, HPSE could be a potential link between MPX with SARS-CoV-2 in Covid-19 era.
Collapse
Affiliation(s)
- Hayder M. Al-Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, ALmustansiriyiah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, ALmustansiriyiah University, Baghdad, Iraq
| | - Helal F. Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, 71515 Egypt
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770 Australia
- AFNP Med, 1030 Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283 Wuppertal, Germany
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Damanhour, 22511 Egypt
| |
Collapse
|
20
|
Lebsir N, Zoulim F, Grigorov B. Heparanase-1: From Cancer Biology to a Future Antiviral Target. Viruses 2023; 15:237. [PMID: 36680276 PMCID: PMC9860851 DOI: 10.3390/v15010237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are a major constituent of the extracellular matrix (ECM) and are found to be implicated in viral infections, where they play a role in both cell entry and release for many viruses. The enzyme heparanase-1 is the only known endo-beta-D-glucuronidase capable of degrading heparan sulphate (HS) chains of HSPGs and is thus important for regulating ECM homeostasis. Heparanase-1 expression is tightly regulated as the uncontrolled cleavage of HS may result in abnormal cell activation and significant tissue damage. The overexpression of heparanase-1 correlates with pathological scenarios and is observed in different human malignancies, such as lymphoma, breast, colon, lung, and hepatocellular carcinomas. Interestingly, heparanase-1 has also been documented to be involved in numerous viral infections, e.g., HSV-1, HPV, DENV. Moreover, very recent reports have demonstrated a role of heparanase-1 in HCV and SARS-CoV-2 infections. Due to the undenied pro-carcinogenic role of heparanase-1, multiple inhibitors have been developed, some reaching phase II and III in clinical studies. However, the use of heparanase inhibitors as antivirals has not yet been proposed. If it can be assumed that heparanase-1 is implicated in numerous viral life cycles, its inhibition by specific heparanase-acting compounds should result in a blockage of viral infection. This review addresses the perspectives of using heparanase inhibitors, not only for cancer treatment, but also as antivirals. Eventually, the development of a novel class antivirals targeting a cellular protein could help to alleviate the resistance problems seen with some current antiretroviral therapies.
Collapse
Affiliation(s)
- Nadjet Lebsir
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69434 Lyon, France
- Confluence: Sciences et Humanités (EA 1598), UCLy, 10 Place des Archives, 69002 Lyon, France
| | - Fabien Zoulim
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69434 Lyon, France
- Hospices Civils de Lyon, 69002 Lyon, France
| | - Boyan Grigorov
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69434 Lyon, France
| |
Collapse
|
21
|
Elste J, Chan A, Patil C, Tripathi V, Shadrack DM, Jaishankar D, Hawkey A, Mungerson MS, Shukla D, Tiwari V. Archaic connectivity between the sulfated heparan sulfate and the herpesviruses - An evolutionary potential for cross-species interactions. Comput Struct Biotechnol J 2023; 21:1030-1040. [PMID: 36733705 PMCID: PMC9880898 DOI: 10.1016/j.csbj.2023.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/04/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023] Open
Abstract
The structural diversity of metazoic heparan sulfate (HS) composed of unique sulfated domains is remarkably preserved among various vertebrates and invertebrate species. Interestingly the sulfated moieties of HS have been known as the key determinants generating extraordinary ligand binding sites in the HS chain to regulate multiple biological functions and homeostasis. One such ligand for 3-O sulfation in the HS chain is a glycoprotein D (gD) from an ancient herpesvirus, herpes simplex virus (HSV). This interaction between gD and 3-O sulfated HS leads to virus-cell fusion to promote HSV entry. It is quite astonishing that HSV-1, which infects two-thirds of the world population, is also capable of causing severe diseases in primates and non-primates including primitive zebrafish. Supporting evidence that HSV may cross the species barrier comes from the fact that an enzymatic modification in HS encoded by 3-O sulfotransferase-3 (3-OST-3) from a vertebrate zoonotic species enhances HSV-1 infectivity. The latter phenomenon suggests the possible role of sulfated-HS as an entry receptor during reverse zoonosis, especially during an event when humans encounter domesticated animals in proximity. In this mini-review, we explore the possibility that structural diversity in HS may have played a substantial role in species-specific adaptability for herpesviruses in general including their potential role in promoting cross-species transmission.
Collapse
Affiliation(s)
- James Elste
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Angelica Chan
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Chandrashekhar Patil
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, IL 60612, USA
| | - Vinisha Tripathi
- Mountain Vista High School, 10585 Mountain Vista Ridge, Highlands Ranch, CO 80126, USA
| | - Daniel M. Shadrack
- Department of Chemistry, Faculty of Natural and Applied Sciences, St John's University of Tanzania, Dodoma, Tanzania
| | - Dinesh Jaishankar
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew Hawkey
- Department of Biomedical Sciences, Midwestern University, Downers Grove, IL 60515, USA
| | - Michelle Swanson Mungerson
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Deepak Shukla
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, IL 60612, USA
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine and College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA,Corresponding author.
| |
Collapse
|
22
|
Petrillo F, Petrillo A, Sasso FP, Schettino A, Maione A, Galdiero M. Viral Infection and Antiviral Treatments in Ocular Pathologies. Microorganisms 2022; 10:2224. [PMID: 36363815 PMCID: PMC9694090 DOI: 10.3390/microorganisms10112224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 08/27/2023] Open
Abstract
Ocular viral infections are common and widespread globally. These infectious diseases are a major cause of acute red eyes and vision loss. The eye and its nearby tissues can be infected by several viral agents, causing infections with a short course and limited ocular implications or a long clinical progression and serious consequences for the function and structure of the ocular region. Several surveillance studies underline the increased emergence of drug resistance among pathogenic viral strains, limiting treatment options for these infections. Currently, in the event of resistant infections, topical or systemic corticosteroids are useful in the management of associated immune reactions in the eye, which contribute to ocular dysfunction. Many cases of viral eye infections are misdiagnosed as being of bacterial origin. In these cases, therapy begins late and is not targeted at the actual cause of the infection, often leading to severe ocular compromises, such as corneal infiltrates, conjunctival scarring, and reduced visual acuity. The present study aims at a better understanding of the viral pathogens that cause eye infections, along with the treatment options available.
Collapse
Affiliation(s)
- Francesco Petrillo
- Azienda Ospedaliera Universitaria-Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | | | | | - Antonietta Schettino
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Angela Maione
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| |
Collapse
|
23
|
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
|
24
|
Gallard C, Lebsir N, Khursheed H, Reungoat E, Plissonnier ML, Bré J, Michelet M, Chouik Y, Zoulim F, Pécheur EI, Bartosch B, Grigorov B. Heparanase-1 is upregulated by hepatitis C virus and favors its replication. J Hepatol 2022; 77:29-41. [PMID: 35085593 DOI: 10.1016/j.jhep.2022.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 01/03/2022] [Accepted: 01/13/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Over time, chronic HCV infection can lead to hepatocellular carcinoma (HCC), a process that involves changes to the liver extracellular matrix (ECM). However, the exact mechanisms by which HCV induces HCC remain unclear. Therefore, we sought to investigate the impact of HCV on the liver ECM, with a focus on heparanase-1 (HPSE). METHODS HPSE expression was assessed by quantitative reverse-transcription PCR, immunoblotting and immunofluorescence in liver biopsies infected or not with HCV, and in 10-day-infected hepatoma Huh7.5 cells. Cell lines deficient for or overexpressing HPSE were established to study its role during infection. RESULTS HCV propagation led to significant HPSE induction, in vivo and in vitro. HPSE enhanced infection when exogenously expressed or supplemented as a recombinant protein. Conversely, when HPSE expression was downregulated or its activity blocked, HCV infection dropped, suggesting a role of HPSE in the HCV life cycle. We further studied the underlying mechanisms of such observations and found that HPSE favored HCV release by enhancing CD63 synthesis and exosome secretion, but not by stimulating HCV entry or genome replication. We also showed that virus-induced oxidative stress was involved in HPSE induction, most likely through NF-κB activation. CONCLUSIONS We report for the first time that HCV infection is favored by HPSE, and upregulates HPSE expression and secretion, which may result in pathogenic alterations of the ECM. LAY SUMMARY Chronic hepatitis C virus (HCV) infection can lead to hepatocellular carcinoma development in a process that involves derangement of the extracellular matrix (ECM). Herein, we show that heparanase-1, a protein involved in ECM degradation and remodeling, favors HCV infection and is upregulated by HCV infection; this upregulation may result in pathogenic alterations of the ECM.
Collapse
Affiliation(s)
- Christophe Gallard
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Nadjet Lebsir
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Hira Khursheed
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Emma Reungoat
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Marie-Laure Plissonnier
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Jennifer Bré
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Maud Michelet
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Yasmina Chouik
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France; Hospices Civils de Lyon, Lyon, France
| | - Fabien Zoulim
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France; Hospices Civils de Lyon, Lyon, France
| | - Eve-Isabelle Pécheur
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France.
| | - Birke Bartosch
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Boyan Grigorov
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France.
| |
Collapse
|
25
|
Richter RP, Ashtekar AR, Zheng L, Pretorius D, Kaushlendra T, Sanderson RD, Gaggar A, Richter JR. Glycocalyx heparan sulfate cleavage promotes endothelial cell angiopoietin-2 expression by impairing shear stress-related AMPK/FoxO1 signaling. JCI Insight 2022; 7:155010. [PMID: 35763350 PMCID: PMC9462499 DOI: 10.1172/jci.insight.155010] [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: 09/14/2021] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Angiopoietin-2 (Ang-2) is a key mediator of vascular disease during sepsis, and elevated plasma levels of Ang-2 are associated with organ injury scores and poor clinical outcomes. We have previously observed that biomarkers of endothelial glycocalyx (EG) damage correlate with plasma Ang-2 levels, suggesting a potential mechanistic linkage between EG injury and Ang-2 expression during states of systemic inflammation. However, the cell signaling mechanisms regulating Ang-2 expression following EG damage are unknown. In the current study, we determined the temporal associations between plasma heparan sulfate (HS) levels as a marker of EG erosion and plasma Ang-2 levels in children with sepsis and in mouse models of sepsis. Secondly, we evaluated the role of shear stress-mediated 5'-adenosine monophosphate-activated protein kinase (AMPK) signaling in Ang-2 expression following enzymatic HS cleavage from the surface of human primary lung microvascular endothelial cells (HLMVEC). We found that plasma HS levels peak prior to plasma Ang-2 levels in children and mice with sepsis. Further, we discovered that impaired AMPK signaling contributes to increased Ang-2 expression following HS cleavage from flow conditioned HLMVECs, establishing a novel paradigm by which Ang-2 may be upregulated during sepsis.
Collapse
Affiliation(s)
- Robert P Richter
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, United States of America
| | - Amit R Ashtekar
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, United States of America
| | - Lei Zheng
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, United States of America
| | - Danielle Pretorius
- Department of Surgery, University of Alabama at Birmingham, Birmingham, United States of America
| | - Tripathi Kaushlendra
- Department of Pathology, University of Alabama at Birmingham, Birmingham, United States of America
| | - Ralph D Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, United States of America
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, United States of America
| | - Jillian R Richter
- Department of Surgery, University of Alabama at Birmingham, Birmingham, United States of America
| |
Collapse
|
26
|
CREB3 Plays an Important Role in HPSE-Facilitated HSV-1 Release in Human Corneal Epithelial Cells. Viruses 2022; 14:v14061171. [PMID: 35746643 PMCID: PMC9227461 DOI: 10.3390/v14061171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 01/16/2023] Open
Abstract
Herpes simplex virus type-1 (HSV-1) exploits several host factors to enhance its replication and release from infected cells. It induces the production of host enzyme heparanase (HPSE) to aid in egress. While the mechanism by which HPSE assists in viral release is well-characterized, other host factors that are recruited along with HPSE for viral release are less well understood. In this study, we identify cyclic-AMP-responsive element-binding protein3 (CREB3) as a key player in HPSE-facilitated HSV-1 egress. When CREB3 is transiently upregulated in human corneal epithelial cells, HSV-1 release from the infected cells is correspondingly enhanced. This activity is linked to HPSE expression such that HPSE-transfected corneal epithelial (HCE) cells more highly express CREB3 than wild-type cells while the cells knocked out for HPSE show very little CREB3 expression. CREB3-transfected HCE cells showed significantly higher export of HPSE upon infection than wild-type cells. Our data suggests that coat protein complex II (COPII), which mediates HPSE trafficking, is also upregulated via a CREB3-dependent pathway during HSV-1 infection. Finally, the co-transfection of CREB3 and HPSE in HCE cells shows the highest viral release compared to either treatment alone, establishing CREB3 as a key player in HPSE-facilitated HSV-1 egress.
Collapse
|
27
|
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
|
28
|
Xiang J, Lu M, Shi M, Cheng X, Kwakwa KA, Davis JL, Su X, Bakewell SJ, Zhang Y, Fontana F, Xu Y, Veis DJ, DiPersio JF, Ratner L, Sanderson RD, Noseda A, Mollah S, Li J, Weilbaecher KN. Heparanase Blockade as a Novel Dual-Targeting Therapy for COVID-19. J Virol 2022; 96:e0005722. [PMID: 35319225 PMCID: PMC9006938 DOI: 10.1128/jvi.00057-22] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/27/2022] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused over 5 million deaths worldwide. Pneumonia and systemic inflammation contribute to its high mortality. Many viruses use heparan sulfate proteoglycans as coreceptors for viral entry, and heparanase (HPSE) is a known regulator of both viral entry and inflammatory cytokines. We evaluated the heparanase inhibitor Roneparstat, a modified heparin with minimum anticoagulant activity, in pathophysiology and therapy for COVID-19. We found that Roneparstat significantly decreased the infectivity of SARS-CoV-2, SARS-CoV-1, and retroviruses (human T-lymphotropic virus 1 [HTLV-1] and HIV-1) in vitro. Single-cell RNA sequencing (scRNA-seq) analysis of cells from the bronchoalveolar lavage fluid of COVID-19 patients revealed a marked increase in HPSE gene expression in CD68+ macrophages compared to healthy controls. Elevated levels of HPSE expression in macrophages correlated with the severity of COVID-19 and the expression of inflammatory cytokine genes, including IL6, TNF, IL1B, and CCL2. In line with this finding, we found a marked induction of HPSE and numerous inflammatory cytokines in human macrophages challenged with SARS-CoV-2 S1 protein. Treatment with Roneparstat significantly attenuated SARS-CoV-2 S1 protein-mediated inflammatory cytokine release from human macrophages, through disruption of NF-κB signaling. HPSE knockdown in a macrophage cell line also showed diminished inflammatory cytokine production during S1 protein challenge. Taken together, this study provides a proof of concept that heparanase is a target for SARS-CoV-2-mediated pathogenesis and that Roneparstat may serve as a dual-targeted therapy to reduce viral infection and inflammation in COVID-19. IMPORTANCE The complex pathogenesis of COVID-19 consists of two major pathological phases: an initial infection phase elicited by SARS-CoV-2 entry and replication and an inflammation phase that could lead to tissue damage, which can evolve into acute respiratory failure or even death. While the development and deployment of vaccines are ongoing, effective therapy for COVID-19 is still urgently needed. In this study, we explored HPSE blockade with Roneparstat, a phase I clinically tested HPSE inhibitor, in the context of COVID-19 pathogenesis. Treatment with Roneparstat showed wide-spectrum anti-infection activities against SARS-CoV-2, HTLV-1, and HIV-1 in vitro. In addition, HPSE blockade with Roneparstat significantly attenuated SARS-CoV-2 S1 protein-induced inflammatory cytokine release from human macrophages through disruption of NF-κB signaling. Together, this study provides a proof of principle for the use of Roneparstat as a dual-targeting therapy for COVID-19 to decrease viral infection and dampen the proinflammatory immune response mediated by macrophages.
Collapse
Affiliation(s)
- Jingyu Xiang
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mijia Lu
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Min Shi
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xiaogang Cheng
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kristin A. Kwakwa
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jennifer L. Davis
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xinming Su
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne J. Bakewell
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yuexiu Zhang
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Francesca Fontana
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yalin Xu
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Deborah J. Veis
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John F. DiPersio
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lee Ratner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ralph D. Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Shamim Mollah
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jianrong Li
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Katherine N. Weilbaecher
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
29
|
Grinage E, Shukla D. Optineurin in ocular herpes infection. Exp Eye Res 2022; 219:109059. [PMID: 35390332 DOI: 10.1016/j.exer.2022.109059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 11/29/2022]
Abstract
Herpes Simplex Virus-1 (HSV-1) is a neurotropic virus that can infect humans in the eye and travel to the trigeminal ganglion to establish latency. HSV-1 causes various disease states in both the primary and secondary sites of infection including the eye and the nervous system. This DNA virus exploits various adaptive measures to infect host cells, hijack host cell proteins, evade host immune response and spread from cell-to-cell to avoid immune detection. Recent data suggest that Optineurin (OPTN), a host protein, is a key restriction factor that prevents cell-to-cell spread of HSV-1 and guards against serious damage to the nervous system during infection. In recent years OPTN has gained increased attention because of its involvement in cellular mechanisms that promote homeostasis and prevent neurodegeneration. At the center of it all is the role OPTN plays as a receptor for selective autophagy. This review summarizes our latest understanding of the viral lifecycle, disease pathologies, and OPTN-mediated protective mechanisms during HSV-1 infection of the eye and the nervous system. We specifically highlight recent discoveries that implicate OPTN as crucial in the prevention of ocular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Earon Grinage
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA; Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA.
| |
Collapse
|
30
|
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
|
31
|
Ostrovsky O, Beider K, Morgulis Y, Bloom N, Cid-Arregui A, Shimoni A, Vlodavsky I, Nagler A. CMV Seropositive Status Increases Heparanase SNPs Regulatory Activity, Risk of Acute GVHD and Yield of CD34 + Cell Mobilization. Cells 2021; 10:cells10123489. [PMID: 34943994 PMCID: PMC8700738 DOI: 10.3390/cells10123489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Heparanase is an endo-β-glucuronidase that is best known for its pro-cancerous effects but is also implicated in the pathogenesis of various viruses. Activation of heparanase is a common strategy to increase viral spread and trigger the subsequent inflammatory cascade. Using a Single Nucleotide Polymorphisms (SNP)-associated approach we identified enhancer and insulator regions that regulate HPSE expression. Although a role for heparanase in viral infection has been noticed, the impact of HPSE functional SNPs has not been determined. We investigated the effect of cytomegalovirus (CMV) serostatus on the involvement of HPSE enhancer and insulator functional SNPs in the risk of acute graft versus host disease (GVHD) and granulocyte-colony stimulating factor related CD34+ mobilization. A significant correlation between the C alleles of insulator rs4364254 and rs4426765 and CMV seropositivity was found in healthy donors and patients with hematological malignancies. The risk of developing acute GVHD after hematopoietic stem cell transplantation was identified only in CMV-seropositive patients. A significant correlation between the enhancer rs4693608 and insulator rs28649799 and CD34+ cell mobilization was demonstrated in the CMV-seropositive donors. It is thus conceivable that latent CMV infection modulates heparanase regulatory regions and enhances the effect of functional SNPs on heparanase function in normal and pathological processes.
Collapse
Affiliation(s)
- Olga Ostrovsky
- Chaim Sheba Medical Center, Department of Hematology and Bone Marrow Transplantation, Tel-Hashomer, Ramat Gan 5266202, Israel; (K.B.); (Y.M.); (N.B.); (A.S.); (A.N.)
- Correspondence: ; Tel.: +972-3-5305770
| | - Katia Beider
- Chaim Sheba Medical Center, Department of Hematology and Bone Marrow Transplantation, Tel-Hashomer, Ramat Gan 5266202, Israel; (K.B.); (Y.M.); (N.B.); (A.S.); (A.N.)
| | - Yan Morgulis
- Chaim Sheba Medical Center, Department of Hematology and Bone Marrow Transplantation, Tel-Hashomer, Ramat Gan 5266202, Israel; (K.B.); (Y.M.); (N.B.); (A.S.); (A.N.)
| | - Nira Bloom
- Chaim Sheba Medical Center, Department of Hematology and Bone Marrow Transplantation, Tel-Hashomer, Ramat Gan 5266202, Israel; (K.B.); (Y.M.); (N.B.); (A.S.); (A.N.)
| | | | - Avichai Shimoni
- Chaim Sheba Medical Center, Department of Hematology and Bone Marrow Transplantation, Tel-Hashomer, Ramat Gan 5266202, Israel; (K.B.); (Y.M.); (N.B.); (A.S.); (A.N.)
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 3525433, Israel;
| | - Arnon Nagler
- Chaim Sheba Medical Center, Department of Hematology and Bone Marrow Transplantation, Tel-Hashomer, Ramat Gan 5266202, Israel; (K.B.); (Y.M.); (N.B.); (A.S.); (A.N.)
| |
Collapse
|
32
|
Heparanase-Induced Activation of AKT Stabilizes β-Catenin and Modulates Wnt/β-Catenin Signaling during Herpes Simplex Virus 1 Infection. mBio 2021; 12:e0279221. [PMID: 34749529 PMCID: PMC8576534 DOI: 10.1128/mbio.02792-21] [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] [Indexed: 01/09/2023] Open
Abstract
Under pathological conditions like herpes simplex virus 1 (HSV-1) infection, host-pathogen interactions lead to major reconstruction of the host protein network, which contributes to the dysregulation of signaling pathways and disease onset. Of note is the upregulation of a multifunctional host protein, heparanase (HPSE), following infection, which serves as a mediator in HSV-1 replication. In this study, we identify a novel function of HPSE and highlight it as a key regulator of β-catenin signal transduction. The regulatory role of HPSE on the activation, nuclear translocation, and signal transduction of β-catenin disrupts cellular homeostasis and establishes a pathogenic environment that promotes viral replication. Under normal physiological conditions, β-catenin is bound to a group of proteins, referred to as the destruction complex, and targeted for ubiquitination and, ultimately, degradation. We show that virus-induced upregulation of HPSE leads to the activation of Akt and subsequent stabilization and activation of β-catenin through (i) the release of β-catenin from the destruction complex, and (ii) direct phosphorylation of β-catenin at Ser552. This study also provides an in-depth characterization of the proviral role of β-catenin signaling during HSV-1 replication using physiologically relevant cell lines and in vivo models of ocular infection. Furthermore, pharmacological inhibitors of this pathway generated a robust antiviral state against multiple laboratory and clinical strains of HSV-1. Collectively, our findings assign a novel regulatory role to HPSE as a driver of β-catenin signaling in HSV-1 infection. IMPORTANCE Heparanase (HPSE) and β-catenin have independently been implicated in regulating key pathophysiological processes, including neovascularization, angiogenesis, and inflammation; however, the relationship between the two proteins has remained elusive thus far. For that reason, characterizing this relationship is crucial and can lead to the development of novel therapeutics. For HSV-1 specifically, current antivirals are not able to abolish the virus from the host, leaving patients susceptible to episodes of viral reactivation. Identifying a host-based intervention can provide a better alternative with enhanced efficacy and sustained relief.
Collapse
|
33
|
Mayfosh AJ, Goodall KJ, Nguyen T, Baschuk N, Hulett MD. Heparanase is a regulator of natural killer cell activation and cytotoxicity. J Leukoc Biol 2021; 111:1211-1224. [PMID: 34693552 DOI: 10.1002/jlb.3a0420-259rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Heparanase is the only mammalian enzyme capable of cleaving heparan sulfate, a glycosaminoglycan of the extracellular matrix and cell surfaces. Most immune cells express heparanase that contributes to a range of functions including cell migration and cytokine expression. Heparanase also promotes natural killer (NK) cell migration; however, its role in other NK cell functions remains to be defined. In this study, heparanase-deficient (Hpse-/- ) mice were used to assess the role of heparanase in NK cell cytotoxicity, activation, and cytokine production. Upon challenge with the immunostimulant polyinosinic:polycytidylic acid (poly(I:C)), NK cells isolated from Hpse-/- mice displayed impaired cytotoxicity against EO771.LMB cells and reduced levels of activation markers CD69 and NKG2D. However, in vitro cytokine stimulation of wild-type and Hpse-/- NK cells resulted in similar CD69 and NKG2D expression, suggesting the impaired NK cell activation in Hpse-/- mice results from elements within the in vivo niche. NK cells are activated in vivo by dendritic cells (DCs) in response to poly(I:C). Poly(I:C)-stimulated Hpse-/- bone marrow DCs (BMDCs) expressed less IL-12, and when cultured with Hpse-/- NK cells, less MCP-1 mRNA and protein was detected. Although cell-cell contact is important for DC-mediated NK cell activation, co-cultures of Hpse-/- BMDCs and NK cells showed similar levels of contact to wild-type cells, suggesting heparanase contributes to NK cell activation independently of cell-cell contact with DCs. These observations define a role for heparanase in NK cell cytotoxicity and activation and have important implications for how heparanase inhibitors currently in clinical trials for metastatic cancer may impact NK cell immunosurveillance.
Collapse
Affiliation(s)
- Alyce J Mayfosh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Katharine J Goodall
- oNKo-innate Pty. Ltd. Monash Biomedicine Discovery Institute, Clayton, Australia
| | - Tien Nguyen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Nikola Baschuk
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
- Heart Regeneration Group, Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| |
Collapse
|
34
|
Mayfosh AJ, Nguyen TK, Hulett MD. The Heparanase Regulatory Network in Health and Disease. Int J Mol Sci 2021; 22:11096. [PMID: 34681753 PMCID: PMC8541136 DOI: 10.3390/ijms222011096] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) is a structural framework that has many important physiological functions which include maintaining tissue structure and integrity, serving as a barrier to invading pathogens, and acting as a reservoir for bioactive molecules. This cellular scaffold is made up of various types of macromolecules including heparan sulfate proteoglycans (HSPGs). HSPGs comprise a protein core linked to the complex glycosaminoglycan heparan sulfate (HS), the remodeling of which is important for many physiological processes such as wound healing as well as pathological processes including cancer metastasis. Turnover of HS is tightly regulated by a single enzyme capable of cleaving HS side chains: heparanase. Heparanase upregulation has been identified in many inflammatory diseases including atherosclerosis, fibrosis, and cancer, where it has been shown to play multiple roles in processes such as epithelial-mesenchymal transition, angiogenesis, and cancer metastasis. Heparanase expression and activity are tightly regulated. Understanding the regulation of heparanase and its downstream targets is attractive for the development of treatments for these diseases. This review provides a comprehensive overview of the regulators of heparanase as well as the enzyme's downstream gene and protein targets, and implications for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Alyce J. Mayfosh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| | - Tien K. Nguyen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| | - Mark D. Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| |
Collapse
|
35
|
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
|
36
|
Dong H, Wang Z, Zhao D, Leng X, Zhao Y. Antiviral strategies targeting herpesviruses. J Virus Erad 2021; 7:100047. [PMID: 34141443 PMCID: PMC8187247 DOI: 10.1016/j.jve.2021.100047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 11/22/2022] Open
Abstract
Herpesviruses, known as large DNA viruses, have a wide host range. In addition to human beings, cattle, and horses, even carp can be hosts for herpesvirus infection. Herpesviruses are pathogens possessing elaborate mechanisms that regulate host cell components for its replication, assembly and generating mature virus particles that can infect humans and most animals, usually causing multiple and lifelong infections. In addition, several human diseases, such as genital or mouth herpes, mononucleosis, and Burkitt lymphoma, are usually associated with herpesvirus infection. Blocking the steps of viral infection, such as entry, replication and assembly, may be an effective way for many different herpes viruses and their related diseases. Therefore, we aim to describe antiviral agents that are able to prevent herpesvirus entry, replication and assembly in host cells. We summarize antiviral strategies, including certain small molecular drugs, RNA interference and CRISPR/Cas9 system-based antiviral approaches, which represent promising approaches.
Collapse
Affiliation(s)
| | | | - Daqing Zhao
- Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyang Leng
- Changchun University of Chinese Medicine, Changchun, China
| | - Yicheng Zhao
- Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
37
|
Agelidis A, Turturice BA, Suryawanshi RK, Yadavalli T, Jaishankar D, Ames J, Hopkins J, Koujah L, Patil CD, Hadigal SR, Kyzar EJ, Campeau A, Wozniak JM, Gonzalez DJ, Vlodavsky I, Li JP, Perkins DL, Finn PW, Shukla D. Disruption of innate defense responses by endoglycosidase HPSE promotes cell survival. JCI Insight 2021; 6:144255. [PMID: 33621216 PMCID: PMC8119219 DOI: 10.1172/jci.insight.144255] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/18/2021] [Indexed: 01/03/2023] Open
Abstract
The drive to withstand environmental stresses and defend against invasion is a universal trait extant in all forms of life. While numerous canonical signaling cascades have been characterized in detail, it remains unclear how these pathways interface to generate coordinated responses to diverse stimuli. To dissect these connections, we followed heparanase (HPSE), a protein best known for its endoglycosidic activity at the extracellular matrix but recently recognized to drive various forms of late-stage disease through unknown mechanisms. Using herpes simplex virus-1 (HSV-1) infection as a model cellular perturbation, we demonstrate that HPSE acts beyond its established enzymatic role to restrict multiple forms of cell-intrinsic defense and facilitate host cell reprogramming by the invading pathogen. We reveal that cells devoid of HPSE are innately resistant to infection and counteract viral takeover through multiple amplified defense mechanisms. With a unique grasp of the fundamental processes of transcriptional regulation and cell death, HPSE represents a potent cellular intersection with broad therapeutic potential.
Collapse
Affiliation(s)
- Alex Agelidis
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | - Benjamin A. Turturice
- Department of Microbiology and Immunology
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | - Dinesh Jaishankar
- Department of Ophthalmology and Visual Sciences, and
- Department of Dermatology, Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Joshua Ames
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | - James Hopkins
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | - Lulia Koujah
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | | | | | - Evan J. Kyzar
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Anaamika Campeau
- Department of Pharmacology and
- Skaggs School of Pharmacy, UCSD, San Diego, La Jolla, California, USA
| | - Jacob M. Wozniak
- Department of Pharmacology and
- Skaggs School of Pharmacy, UCSD, San Diego, La Jolla, California, USA
| | - David J. Gonzalez
- Department of Pharmacology and
- Skaggs School of Pharmacy, UCSD, San Diego, La Jolla, California, USA
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Jin-ping Li
- Department of Medical Biochemistry and Microbiology, University of Uppsala, Uppsala, Sweden
| | - David L. Perkins
- Division of Nephrology, Department of Medicine, and
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Patricia W. Finn
- Department of Microbiology and Immunology
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Calabretta E, Moraleda JM, Iacobelli M, Jara R, Vlodavsky I, O’Gorman P, Pagliuca A, Mo C, Baron RM, Aghemo A, Soiffer R, Fareed J, Carlo‐Stella C, Richardson P. COVID-19-induced endotheliitis: emerging evidence and possible therapeutic strategies. Br J Haematol 2021; 193:43-51. [PMID: 33538335 PMCID: PMC8014053 DOI: 10.1111/bjh.17240] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/19/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Eleonora Calabretta
- Department of Oncology and HematologyHumanitas Cancer CenterHumanitas Clinical and Research Center ‐ IRCCSRozzanoMilanItaly
- Department of Biomedical SciencesHumanitas UniversityRozzanoMilanItaly
| | - Jose M. Moraleda
- Department of HematologyStem Cell Transplant and Cell Therapy UnitVirgen de la Arrixaca University HospitalIMIB‐ArrixacaUniversity of MurciaMurciaSpain
| | | | - Ruben Jara
- Department of Critical Care MedicineVirgen de la Arrixaca University HospitalIMIB‐ArrixacaUniversity of MurciaMurciaSpain
| | - Israel Vlodavsky
- The Rappaport Faculty of MedicineTechnion Integrated Cancer Center (TICC)HaifaIsrael
| | - Peter O’Gorman
- Haematology DepartmentMater Misericordiae University HospitalDublinIreland
| | - Antonio Pagliuca
- Department of HaematologyKings College Hospital NHS Foundation TrustLondonUK
| | - Clifton Mo
- Department of Medical OncologyDivision of Hematologic MalignanciesDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMAUSA
| | - Rebecca M. Baron
- Division of Pulmonary and Critical Care MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Alessio Aghemo
- Division of Hepatology and Internal MedicineHumanitas Clinical and Research Center ‐ IRCCSMilanItaly
- Department of Biomedical SciencesHumanitas UniversityRozzanoMilanItaly
| | - Robert Soiffer
- Department of Medical OncologyDivision of Hematologic MalignanciesDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMAUSA
| | | | - Carmelo Carlo‐Stella
- Department of Oncology and HematologyHumanitas Cancer CenterHumanitas Clinical and Research Center ‐ IRCCSRozzanoMilanItaly
- Department of Biomedical SciencesHumanitas UniversityRozzanoMilanItaly
| | - Paul Richardson
- Department of Medical OncologyDivision of Hematologic MalignanciesDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
41
|
Gerlza T, Trojacher C, Kitic N, Adage T, Kungl AJ. Development of Molecules Antagonizing Heparan Sulfate Proteoglycans. Semin Thromb Hemost 2021; 47:316-332. [PMID: 33794555 DOI: 10.1055/s-0041-1725067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) occur in almost every tissue of the human body and consist of a protein core, with covalently attached glycosaminoglycan polysaccharide chains. These glycosaminoglycans are characterized by their polyanionic nature, due to sulfate and carboxyl groups, which are distributed along the chain. These chains can be modified by different enzymes at varying positions, which leads to huge diversity of possible structures with the complexity further increased by varying chain lengths. According to their location, HSPGs are divided into different families, the membrane bound, the secreted extracellular matrix, and the secretory vesicle family. As members of the extracellular matrix, they take part in cell-cell communication processes on many levels and with different degrees of involvement. Of particular therapeutic interest is their role in cancer and inflammation as well as in infectious diseases. In this review, we give an overview of the current status of medical approaches to antagonize HSPG function in pathology.
Collapse
Affiliation(s)
- Tanja Gerlza
- Karl-Franzens University Graz, Institute of Pharmaceutical Sciences, Graz, Austria
| | - Christina Trojacher
- Karl-Franzens University Graz, Institute of Pharmaceutical Sciences, Graz, Austria
| | - Nikola Kitic
- Karl-Franzens University Graz, Institute of Pharmaceutical Sciences, Graz, Austria
| | | | - Andreas J Kungl
- Karl-Franzens University Graz, Institute of Pharmaceutical Sciences, Graz, Austria.,Antagonis Biotherapeutics GmbH, Graz, Austria
| |
Collapse
|
42
|
Koujah L, Allaham M, Patil CD, Ames JM, Suryawanshi RK, Yadavalli T, Agelidis A, Mun C, Surenkhuu B, Jain S, Shukla D. Entry receptor bias in evolutionarily distant HSV-1 clinical strains drives divergent ocular and nervous system pathologies. Ocul Surf 2021; 21:238-249. [PMID: 33766740 DOI: 10.1016/j.jtos.2021.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 01/13/2023]
Abstract
PURPOSE Herpes simplex virus-1 (HSV-1) infection leads to varying pathologies including the development of ocular lesions, stromal keratitis and encephalitis. While the role for host immunity in disease progression is well understood, the contribution of genetic variances in generating preferential viral entry receptor usage and resulting immunopathogenesis in humans are not known. METHODS Ocular cultures were obtained from patients presenting distinct pathologies of herpes simplex keratitis (HSK). Next-generation sequencing and subsequent analysis characterized genetic variances among the strains and estimated evolutionary divergence. Murine model of ocular infection was used to assess phenotypic contributions of strain variances on damage to the ocular surface and propagation of innate immunity. Flow cytometry of eye tissue identified differential recruitment of immune cell populations, cytokine array probed for programming of local immune response in the draining lymph node and histology was used to assess inflammation of the trigeminal ganglion (TG). Ex-vivo corneal cultures and in-vitro studies elucidated the role of genetic variances in altering host-pathogen interactions, leading to divergent host responses. RESULTS Phylogenetic analysis of the clinical isolates suggests evolutionary divergence among currently circulating HSV-1 strains. Mutations causing alterations in functional host interactions were identified, particularly in viral entry glycoproteins which generated a receptor bias to herpesvirus entry mediator, an immune modulator involved in immunopathogenic diseases like HSK, leading to exacerbated ocular surface pathologies and heightened viral burden in the TG and brainstem. CONCLUSIONS Our data suggests receptor bias resulting from genetic variances in clinical strains may dictate disease severity and treatment outcome.
Collapse
Affiliation(s)
- Lulia Koujah
- 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
| | - Mowafak Allaham
- Department of Mathematics, Statistics and Computer Science, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Chandrashekhar D Patil
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Joshua M Ames
- 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
| | - Rahul K Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Alex Agelidis
- 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
| | - Christine Mun
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bayasgalan Surenkhuu
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sandeep Jain
- Department of Ophthalmology and Visual Sciences, 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
|
43
|
Agelidis A, Suryawanshi RK, Patil CD, Campeau A, Gonzalez DJ, Shukla D. Dissociation of DNA damage sensing by endoglycosidase HPSE. iScience 2021; 24:102242. [PMID: 33748723 PMCID: PMC7957091 DOI: 10.1016/j.isci.2021.102242] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/09/2021] [Accepted: 02/24/2021] [Indexed: 12/25/2022] Open
Abstract
Balance between cell proliferation and elimination is critical in handling threats both exogenous and of internal dysfunction. Recent work has implicated a conserved but poorly understood endoglycosidase heparanase (HPSE) in the restriction of innate defense responses, yet biochemical mediators of these key functions remained unclear. Here, an unbiased immunopurification proteomics strategy is employed to identify and rank uncharacterized interactions between HPSE and mediators of canonical signaling pathways linking cell cycle and stress responses. We demonstrate with models of genotoxic stress including herpes simplex virus infection and chemotherapeutic treatment that HPSE dampens innate responses to double-stranded DNA breakage by interfering with signal transduction between initial sensors and downstream mediators. Given the long-standing recognition of HPSE in driving late-stage inflammatory disease exemplified by tissue destruction and cancer metastasis, modulation of this protein with control over the DNA damage response imparts a unique strategy in the development of unconventional multivalent therapy. HPSE binds key proteins at interface of DNA damage signaling and IFN responses Nuclear translocation of DNA damage transducer ATM is enhanced in absence of HPSE Cells lacking HPSE display enhanced sensitivity to DNA damage-induced death HPSE interfaces with regulators of DNA damage response to influence cell fate
Collapse
Affiliation(s)
- Alex Agelidis
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rahul K. Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Chandrashekhar D. Patil
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Anaamika Campeau
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy, University of California, San Diego, La Jolla, CA 92093, USA
| | - David J. Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy, University of California, San Diego, La Jolla, CA 92093, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Corresponding author
| |
Collapse
|
44
|
Madavaraju K, Koganti R, Volety I, Yadavalli T, Shukla D. Herpes Simplex Virus Cell Entry Mechanisms: An Update. Front Cell Infect Microbiol 2021; 10:617578. [PMID: 33537244 PMCID: PMC7848091 DOI: 10.3389/fcimb.2020.617578] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus (HSV) can infect a broad host range and cause mild to life threating infections in humans. The surface glycoproteins of HSV are evolutionarily conserved and show an extraordinary ability to bind more than one receptor on the host cell surface. Following attachment, the virus fuses its lipid envelope with the host cell membrane and releases its nucleocapsid along with tegument proteins into the cytosol. With the help of tegument proteins and host cell factors, the nucleocapsid is then docked into the nuclear pore. The viral double stranded DNA is then released into the host cell’s nucleus. Released viral DNA either replicates rapidly (more commonly in non-neuronal cells) or stays latent inside the nucleus (in sensory neurons). The fusion of the viral envelope with host cell membrane is a key step. Blocking this step can prevent entry of HSV into the host cell and the subsequent interactions that ultimately lead to production of viral progeny and cell death or latency. In this review, we have discussed viral entry mechanisms including the pH-independent as well as pH-dependent endocytic entry, cell to cell spread of HSV and use of viral glycoproteins as an antiviral target.
Collapse
Affiliation(s)
- Krishnaraju Madavaraju
- Shukla Lab, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Raghuram Koganti
- Shukla Lab, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Ipsita Volety
- Shukla Lab, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Tejabhiram Yadavalli
- Shukla Lab, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Deepak Shukla
- Shukla Lab, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States.,Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
45
|
Yadavalli T, Suryawanshi R, Koganti R, Hopkins J, Ames J, Koujah L, Iqbal A, Madavaraju K, Agelidis A, Shukla D. Standalone or combinatorial phenylbutyrate therapy shows excellent antiviral activity and mimics CREB3 silencing. SCIENCE ADVANCES 2020; 6:eabd9443. [PMID: 33277262 PMCID: PMC7821892 DOI: 10.1126/sciadv.abd9443] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/22/2020] [Indexed: 05/12/2023]
Abstract
Herpesviruses are ubiquitous human pathogens that tightly regulate many cellular pathways including the unfolded protein response to endoplasmic reticulum (ER) stress. Pharmacological modulation of this pathway results in the inhibition of viral replication. In this study, we tested 4-phenylbutyrate (PBA), a chemical chaperone-based potent alleviator of ER stress, for its effects on herpes simplex virus (HSV) type 1 infection. Through in vitro studies, we observed that application of PBA to HSV-infected cells results in the down-regulation of a proviral, ER-localized host protein CREB3 and a resultant inhibition of viral protein synthesis. PBA treatment caused viral inhibition in cultured human corneas and human skin grafts as well as murine models of ocular and genital HSV infection. Thus, we propose that this drug can provide an alternative to current antivirals to treat both ocular HSV-1 and genital HSV-2 infections and may be a strong candidate for human trials.
Collapse
Affiliation(s)
- Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rahul Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - James Hopkins
- 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
| | - Joshua Ames
- 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
| | - Lulia Koujah
- 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
| | - Aqsa Iqbal
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Krishnaraju Madavaraju
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Alex Agelidis
- Department of Ophthalmology and Visual Sciences, 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
|
46
|
Koganti R, Suryawanshi R, Shukla D. Heparanase, cell signaling, and viral infections. Cell Mol Life Sci 2020; 77:5059-5077. [PMID: 32462405 PMCID: PMC7252873 DOI: 10.1007/s00018-020-03559-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Abstract
Heparanase (HPSE) is a multifunctional protein endowed with many non-enzymatic functions and a unique enzymatic activity as an endo-β-D-glucuronidase. The latter allows it to serve as a key modulator of extracellular matrix (ECM) via a well-regulated cleavage of heparan sulfate side chains of proteoglycans at cell surfaces. The cleavage and associated changes at the ECM cause release of multiple signaling molecules with important cellular and pathological functions. New and emerging data suggest that both enzymatic as well as non-enzymatic functions of HPSE are important for health and illnesses including viral infections and virally induced cancers. This review summarizes recent findings on the roles of HPSE in activation, inhibition, or bioavailability of key signaling molecules such as AKT, VEGF, MAPK-ERK, and EGFR, which are known regulators of common viral infections in immune and non-immune cell types. Altogether, our review provides a unique overview of HPSE in cell-survival signaling pathways and how they relate to viral infections.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Rahul Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA.
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
47
|
Shi C, Wang C, Wang H, Yang C, Cai F, Zeng F, Cheng F, Liu Y, Zhou T, Deng B, Vlodavsky I, Li JP, Zhang Y. The Potential of Low Molecular Weight Heparin to Mitigate Cytokine Storm in Severe COVID-19 Patients: A Retrospective Cohort Study. Clin Transl Sci 2020; 13:1087-1095. [PMID: 32881340 PMCID: PMC7719364 DOI: 10.1111/cts.12880] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
On March 11, 2020, the World Health Organization declared its assessment of coronavirus disease 2019 (COVID-19) as a global pandemic. However, specific anti-severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) drugs are still under development, and patients are managed by multiple complementary treatments. We performed a retrospective analysis to compare and evaluate the effect of low molecular weight heparin (LMWH) treatment on disease progression. For this purpose, the clinical records and laboratory indicators were extracted from electronic medical records of 42 patients with COVID-19 (21 of whom were treated with LMWH, and 21 without LMWH) hospitalized (Union Hospital of Huazhong University of Science and Technology) from February 1 to March 15, 2020. Changes in the percentage of lymphocytes before and after LMWH treatment were significantly different from those in the control group (P = 0.011). Likewise, changes in the levels of D-dimer and fibrinogen degradation products in the LMWH group before and after treatment were significantly different from those in the control group (P = 0.035). Remarkably, IL-6 levels were significantly reduced after LMWH treatment (P = 0.006), indicating that, besides other beneficial properties, LMWH may exert an anti-inflammatory effect and attenuate in part the "cytokine storm" induced by the virus. Our results support the use of LMWH as a potential therapeutic drug for the treatment of COVID-19, paving the way for a subsequent well-controlled clinical study.
Collapse
Affiliation(s)
- Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Cong Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hanxiang Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Yang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zeng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Cheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yihui Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Taotao Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Deng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Israel
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| |
Collapse
|
48
|
Amin R, Tripathi K, Sanderson RD. Nuclear Heparanase Regulates Chromatin Remodeling, Gene Expression and PTEN Tumor Suppressor Function. Cells 2020; 9:cells9092038. [PMID: 32899927 PMCID: PMC7564302 DOI: 10.3390/cells9092038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 02/08/2023] Open
Abstract
Heparanase (HPSE) is an endoglycosidase that cleaves heparan sulfate and has been shown in various cancers to promote metastasis, angiogenesis, osteolysis, and chemoresistance. Although heparanase is thought to act predominantly extracellularly or within the cytoplasm, it is also present in the nucleus, where it may function in regulating gene transcription. Using myeloma cell lines, we report here that heparanase enhances chromatin accessibility and confirm a previous report that it also upregulates the acetylation of histones. Employing the Multiple Myeloma Research Foundation CoMMpass database, we demonstrate that patients expressing high levels of heparanase display elevated expression of proteins involved in chromatin remodeling and several oncogenic factors compared to patients expressing low levels of heparanase. These signatures were consistent with the known function of heparanase in driving tumor progression. Chromatin opening and downstream target genes were abrogated by inhibition of heparanase. Enhanced levels of heparanase in myeloma cells led to a dramatic increase in phosphorylation of PTEN, an event known to stabilize PTEN, leading to its inactivity and loss of tumor suppressor function. Collectively, this study demonstrates that heparanase promotes chromatin opening and transcriptional activity, some of which likely is through its impact on diminishing PTEN tumor suppressor activity.
Collapse
|
49
|
Khanna M, Parish CR. Heparanase: Historical Aspects and Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:71-96. [PMID: 32274707 DOI: 10.1007/978-3-030-34521-1_3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Heparanase is an endo-β-glucuronidase that cleaves at a limited number of internal sites the glycosaminoglycan heparan sulfate (HS). Heparanase enzymatic activity was first reported in 1975 and by 1983 evidence was beginning to emerge that the enzyme was a facilitator of tumor metastasis by cleaving HS chains present in blood vessel basement membranes and, thereby, aiding the passage of tumor cells through blood vessel walls. Due to a range of technical difficulties, it took another 16 years before heparanase was cloned and characterized in 1999 and a further 14 years before the crystal structure of the enzyme was solved. Despite these substantial deficiencies, there was steady progress in our understanding of heparanase long before the enzyme was fully characterized. For example, it was found as early as 1984 that activated T cells upregulate heparanase expression, like metastatic tumor cells, and the enzyme aids the entry of T cells and other leukocytes into inflammatory sites. Furthermore, it was discovered in 1989 that heparanase releases pre-existing growth factors and cytokines associated with HS in the extracellular matrix (ECM), the liberated growth factors/cytokines enhancing angiogenesis and wound healing. There were also the first hints that heparanase may have functions other than enzymatic activity, in 1995 it being reported that under certain conditions the enzyme could act as a cell adhesion molecule. Also, in the same year PI-88 (Muparfostat), the first heparanase inhibitor to reach and successfully complete a Phase III clinical trial was patented.Nevertheless, the cloning of heparanase (also known as heparanase-1) in 1999 gave the field an enormous boost and some surprises. The biggest surprise was that there is only one heparanase encoding gene in the mammalian genome, despite earlier research, based on substrate specificity, suggesting that there are at least three different heparanases. This surprising conclusion has remained unchanged for the last 20 years. It also became evident that heparanase is a family 79 glycoside hydrolase that is initially produced as a pro-enzyme that needs to be processed by proteases to form an enzymatically active heterodimer. A related molecule, heparanase-2, was also discovered that is enzymatically inactive but, remarkably, recently has been shown to inhibit heparanase-1 activity as well as acting as a tumor suppressor that counteracts many of the pro-tumor properties of heparanase-1.The early claim that heparanase plays a key role in tumor metastasis, angiogenesis and inflammation has been confirmed by many studies over the last 20 years. In fact, heparanase expression is enhanced in all major cancer types, namely carcinomas, sarcomas, and hematological malignancies, and correlates with increased metastasis and poor prognosis. Also, there is mounting evidence that heparanase plays a central role in the induction of inflammation-associated cancers. The enzymatic activity of heparanase has also emerged in unexpected situations, such as in the spread of HS-binding viruses and in Type-1 diabetes where the destruction of intracellular HS in pancreatic insulin-producing beta cells precipitates diabetes. But the most extraordinary recent discoveries have been with the realization that heparanase can exert a range of biological activities that are independent of its enzymatic function, most notably activation of several signaling pathways and being a transcription factor that controls methylation of histone tails. Collectively, these data indicate that heparanase is a truly multifunctional protein that has the additional property of cleaving HS chains and releasing from ECM and cell surfaces hundreds of HS-binding proteins with a plethora of functional consequences. Clearly, there are many unique features of this intriguing molecule that still remain to be explored and are highlighted in this Chapter.
Collapse
Affiliation(s)
- Mayank Khanna
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| |
Collapse
|
50
|
Heparanase: Cloning, Function and Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:189-229. [PMID: 32274711 DOI: 10.1007/978-3-030-34521-1_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In 2019, we mark the 20th anniversary of the cloning of the human heparanase gene. Heparanase remains the only known enzyme to cleave heparan sulfate, which is an abundant component of the extracellular matrix. Thus, elucidating the mechanisms underlying heparanase expression and activity is critical to understanding its role in healthy and pathological settings. This chapter provides a historical account of the race to clone the human heparanase gene, describes the intracellular and extracellular function of the enzyme, and explores the various mechanisms regulating heparanase expression and activity at the gene, transcript, and protein level.
Collapse
|