Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Viruses ; 13(12)2021 12 14.
Article in English | MEDLINE | ID: mdl-34960773

ABSTRACT

Notable among the many communicable agents known to infect the human cornea is the human adenovirus, with less than ten adenoviruses having corneal tropism out of more than 100 known types. The syndrome of epidemic keratoconjunctivitis (EKC), caused principally by human adenovirus, presents acutely with epithelial keratitis, and later with stromal keratitis that can be chronic and recurrent. In this review, we discuss the current state of knowledge regarding the molecular biology of adenovirus infection of corneal stromal cells, among which the fibroblast-like keratocyte is the most predominant, in order to elucidate basic pathophysiologic mechanisms of stromal keratitis in the human patient with EKC.


Subject(s)
Adenoviruses, Human/physiology , Cornea/virology , Keratitis/etiology , Adenoviruses, Human/classification , Animals , Cornea/cytology , Cornea/embryology , Host Microbial Interactions , Humans , Interleukin-8/genetics , Keratoconjunctivitis/etiology , Organogenesis , Stromal Cells/virology
2.
Am J Surg Pathol ; 45(10): 1357-1363, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34324455

ABSTRACT

Herpes viruses are known for infecting epithelial cells and manifesting as vesicles. However, herpes viruses can also infect stromal cells. While established in the ocular setting, cutaneous stromal herpes (deep herpes) is previously unreported and may evade clinical and microscopic detection. We searched for skin biopsies with herpes stromal disease. Clinical information was retrieved via electronic medical records and pathology records system. Hematoxylin and eosin slides, immunohistochemical staining, and polymerase chain reaction detection of viral DNA was performed. We identified 12 specimens from 10 patients with cutaneous stromal herpes simplex virus 1/2 (n=7) or varicella-zoster virus infection (n=5). The most common site involved was the buttocks/perianal region (n=6). Ulceration was a frequent dermatologic finding (n=8). Pyoderma gangrenosum was clinically suspected in 6 specimens (50%). Eight patients (80%) were immunosuppressed. Biopsies frequently demonstrated a dense dermal mixed inflammatory infiltrate with subcutaneous extension and enlarged cells with viral cytopathic changes confirmed by herpes simplex virus 1/2 or varicella-zoster virus immunohistochemistry (n=10) or polymerase chain reaction (n=2). Most specimens (67%) lacked evidence of characteristic epidermal keratinocyte infection. This study presents the first known report of the ability of herpes virus to infect deep stromal cells of the dermis. We raise awareness of cutaneous stromal herpes in patients presenting with atypical clinical lesions, particularly while immunocompromised. Establishing the correct diagnosis is critical for initiating therapy.


Subject(s)
Dermis/virology , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/pathogenicity , Herpesvirus 3, Human/pathogenicity , Stromal Cells/virology , Varicella Zoster Virus Infection/virology , Adolescent , Adult , Aged , Antiviral Agents/therapeutic use , DNA, Viral/genetics , Dermis/drug effects , Dermis/pathology , Female , Herpes Simplex/diagnosis , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/drug effects , Herpesvirus 2, Human/genetics , Herpesvirus 3, Human/drug effects , Herpesvirus 3, Human/genetics , Host-Pathogen Interactions , Humans , Male , Middle Aged , Retrospective Studies , Stromal Cells/drug effects , Stromal Cells/pathology , Treatment Outcome , Varicella Zoster Virus Infection/diagnosis , Varicella Zoster Virus Infection/drug therapy , Young Adult
3.
Cardiovasc Res ; 117(6): 1557-1566, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33705542

ABSTRACT

AIMS: Patients with severe respiratory syndrome caused by SARS-CoV-2 undergo cardiac complications due to hyper-inflammatory conditions. Although the presence of the virus has been detected in the myocardium of infected patients, and infection of induced pluripotent cell-derived cardiomyocytes has been demonstrated, the reported expression of Angiotensin-Converting Enzyme-2 (ACE2) in cardiac stromal cells suggests that SARS-CoV-2 may determine cardiac injury by sustaining productive infection and increasing inflammation. METHODS AND RESULTS: We analysed expression of ACE2 receptor in primary human cardiac stromal cells derived from cardiospheres, using proteomics and transcriptomics before exposing them to SARS-CoV-2 in vitro. Using conventional and high sensitivity PCR methods, we measured virus release in the cellular supernatants and monitored the intracellular viral bioprocessing. We performed high-resolution imaging to show the sites of intracellular viral production and demonstrated the presence of viral particles in the cells with electron microscopy. We finally used RT-qPCR assays to detect genes linked to innate immunity and fibrotic pathways coherently regulated in cells after exposure to the virus. CONCLUSIONS: Our findings indicate that cardiac stromal cells are susceptible to SARS-CoV-2 infection and produce variable viral yields depending on the extent of cellular ACE2 receptor expression. Interestingly, these cells also evolved towards hyper-inflammatory/pro-fibrotic phenotypes independently of ACE2 levels. Thus, SARS-CoV-2 infection of myocardial stromal cells could be involved in cardiac injury and explain the high number of complications observed in severe cases of COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Heart Diseases/virology , Myocardium/enzymology , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Stromal Cells/virology , Virion/pathogenicity , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/complications , Chlorocebus aethiops , Female , Fibrosis , Heart Diseases/enzymology , Heart Diseases/pathology , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Myocardium/ultrastructure , Phenotype , Receptors, Virus/genetics , SARS-CoV-2/ultrastructure , Spheroids, Cellular , Stromal Cells/enzymology , Stromal Cells/ultrastructure , Vero Cells , Virion/ultrastructure
4.
Acta Neuropathol Commun ; 8(1): 221, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33308315

ABSTRACT

Cancer-associated fibroblasts (CAFs) are activated fibroblasts constituting the major stromal components in many types of cancer. CAFs contribute to hallmarks of cancer such as proliferation, invasion and immunosuppressive tumor microenvironment, and are associated with poor prognosis of patients with cancer. However, in glioblastoma (GBM), the most common and aggressive primary malignant brain tumor, our knowledge about CAFs or CAF-like stromal cells is limited. Here, using commonly accepted CAF markers, we characterized CAF-like cell populations in clinical glioma specimens and datasets along with mouse models of GBM. We found that tumor-associated pericytes marked by co-expression of fibroblast activation protein α (FAP) and PDGFRß represent major stromal cell subsets in both human GBM and mouse GBM models, while a fraction of mesenchymal neoplastic cells also express FAP in patient tumors. Since oncolytic viruses can kill cancer cells and simultaneously modulate the tumor microenvironment by impacting non-neoplastic populations such as immune cells and tumor vasculature, we further investigated the ability of oncolytic viruses to target GBM-associated stromal cells. An oncolytic adenovirus, ICOVIR15, carrying ∆24-E1A and an RGD-fiber, infects and depletes FAP+ pericytes as well as GBM cells in murine GBM. Our study thus identifies FAP+/PDGFRß+ pericytes as a major CAF-like stromal cell population in GBM, and highlights the unique property of this oncolytic adenovirus to target both GBM cells and GBM-associated stromal FAP+ cells.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Endopeptidases/metabolism , Glioblastoma/metabolism , Membrane Proteins/metabolism , Oncolytic Viruses , Pericytes/metabolism , Animals , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/virology , Disease Models, Animal , Glioblastoma/pathology , Humans , Mice , Oncolytic Virotherapy , Pericytes/cytology , Pericytes/virology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/virology , Tumor Microenvironment
5.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641474

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that can cause severe clinical disease in allograft recipients and infants infected in utero Virus-neutralizing antibodies defined in vitro have been proposed to confer protection against HCMV infection, and the virion envelope glycoprotein B (gB) serves as a major target of neutralizing antibodies. The viral fusion protein gB is nonfusogenic on its own and requires glycoproteins H (gH) and L (gL) for membrane fusion, which is in contrast to requirements of related class III fusion proteins, including vesicular stomatitis virus glycoprotein G (VSV-G) or baculovirus gp64. To explore requirements for gB's fusion activity, we generated a set of chimeras composed of gB and VSV-G or gp64, respectively. These gB chimeras were intrinsically fusion active and led to the formation of multinucleated cell syncytia when expressed in the absence of other viral proteins. Utilizing a panel of virus-neutralizing gB-specific monoclonal antibodies (MAbs), we could demonstrate that syncytium formation of the fusogenic gB/VSV-G chimera can be significantly inhibited by only a subset of neutralizing MAbs which target antigenic domain 5 (AD-5) of gB. This observation argues for differential modes of action of neutralizing anti-gB MAbs and suggests that blocking the membrane fusion function of gB could be one mechanism of antibody-mediated virus neutralization. In addition, our data have important implications for the further understanding of the conformation of gB that promotes membrane fusion as well as the identification of structures in AD-5 that could be targeted by antibodies to block this early step in HCMV infection.IMPORTANCE HCMV is a major global health concern, and antiviral chemotherapy remains problematic due to toxicity of available compounds and the emergence of drug-resistant viruses. Thus, an HCMV vaccine represents a priority for both governmental and pharmaceutical research programs. A major obstacle for the development of a vaccine is a lack of knowledge of the nature and specificities of protective immune responses that should be induced by such a vaccine. Glycoprotein B of HCMV is an important target for neutralizing antibodies and, hence, is often included as a component of intervention strategies. By generation of fusion-active gB chimeras, we were able to identify target structures of neutralizing antibodies that potently block gB-induced membrane fusion. This experimental system provides an approach to screen for antibodies that interfere with gB's fusogenic activity. In summary, our data will likely contribute to both rational vaccine design and the development of antibody-based therapies against HCMV.


Subject(s)
Antibodies, Neutralizing/pharmacology , Cytomegalovirus/genetics , Mutant Chimeric Proteins/genetics , Viral Envelope Proteins/genetics , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Antibodies, Viral/pharmacology , Binding Sites , Cell Fusion , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression , Giant Cells/drug effects , Giant Cells/metabolism , Giant Cells/ultrastructure , Giant Cells/virology , HEK293 Cells , Humans , Mice , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/metabolism , Primary Cell Culture , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/virology , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Envelope Proteins/metabolism
6.
mSphere ; 4(4)2019 07 17.
Article in English | MEDLINE | ID: mdl-31315968

ABSTRACT

Jason Bodily works in the field of tumor virology. In this mSphere of Influence article, he reflects on how "Inactivation of Rb in stromal fibroblasts promotes epithelial cell invasion" by Adam Pickard et al. (EMBO J 31:3092-3103, 2012, https://doi.org/10.1038/emboj.2012.153) has impacted his work by making him think about the role of stromal cells in human papillomavirus infections.


Subject(s)
Epithelial Cells/virology , Oncogenes , Papillomaviridae/genetics , Stromal Cells/virology , Humans , Narration , Papillomaviridae/physiology , Papillomavirus Infections/virology
7.
Cell Rep ; 28(1): 245-256.e4, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31269444

ABSTRACT

Type I interferon (IFNα/ß) pathways are fine-tuned to elicit antiviral protection while minimizing immunopathology; however, the initiating stimuli, target tissues, and underlying mechanisms are unclear. Using models of physiological and dysregulated IFNα/ß receptor (IFNAR1) surface expression, we show here that IFNAR1-dependent signals set the steady-state IFN signature in both hematopoietic and stromal cells. Increased IFNAR1 levels promote a lung environment refractory to early influenza virus replication by elevating the baseline interferon signature. Commensal microbiota drive the IFN signature specifically in lung stroma, as shown by antibiotic treatment and fecal transplantation. Bone marrow chimera experiments identify lung stromal cells as crucially important for early antiviral immunity and stroma-immune cell interaction for late antiviral resistance. We propose that the microbiota-driven interferon signature in lung epithelia impedes early virus replication and that IFNAR1 surface levels fine-tune this signature. Our findings highlight the interplay between bacterial and viral exposure, with important implications for antibiotic use.


Subject(s)
Anti-Bacterial Agents/pharmacology , Influenza A virus , Influenza, Human/immunology , Influenza, Human/microbiology , Lung/immunology , Microbiota/immunology , Receptor, Interferon alpha-beta/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line , Chimera/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Fecal Microbiota Transplantation , Gene Expression Regulation, Viral/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , Humans , Influenza A virus/growth & development , Influenza A virus/immunology , Influenza, Human/drug therapy , Influenza, Human/pathology , Interferon Type I/metabolism , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Lung/drug effects , Lung/microbiology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Seq , Receptor, Interferon alpha-beta/genetics , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/microbiology , Stromal Cells/virology
8.
Am J Reprod Immunol ; 82(4): e13174, 2019 10.
Article in English | MEDLINE | ID: mdl-31338899

ABSTRACT

PROBLEM: We first reported human herpesvirus (HHV)-6A DNA presence in 43% of endometrial cells from women with idiopathic infertility, whereas no fertile control women harbored the virus. We investigated the effect of HHV-6A infection on the immunological status of the endometrium. METHOD OF STUDY: Endometrial biopsies, uterine flushing, and whole blood samples were collected from 67 idiopathic infertile women (mid-secretory phase). We analyzed the endometrial immunological status evaluating: (a) the effect of HHV-6A infection on endometrial immune profile analyzing the ratio of interleukin (IL)-15/ fibroblast growth factor-inducible 14 (Fn-14) and IL-18/ TNF-related weak inducer of apoptosis (TWEAK) mRNA as a biomarker of endometrial (e)natural killer activation/maturation, angiogenesis, and Th1/Th2 balance; (b) endometrial receptivity to trophoblasts in endometrial 3D in vitro model; (c) natural killer (NK) cells and T cells percentage and subpopulations by flow cytometry. RESULTS: We confirmed the presence of HHV-6A infection in a 40% of idiopathic infertile women, characterized by an immune profile reflecting eNK cell cytotoxic activation and a decrease in CD4+ CD25+ CD127dim/- regulatory T cells. The co-culture of endometrial epithelial cells with spheroids generated from the extravillous trophoblast-derived cell line JEG3 showed a twofold expansion of spheroids on endometrial epithelial-stromal cells (ESC) culture surface from HHV-6A negative women while no expansion was observed on the surface of ESC from HHV-6A positive women. CONCLUSION: The identification of an effect of HHV-6A infection on endometrial immune status opens new perspectives in idiopathic infertile women care management. In addition, it would be possible to select antiviral therapies as novel, non-hormonal therapeutic approaches to those idiopathic infertile women characterized by the presence of endometrial HHV-6A infection, to increase their pregnancy rate.


Subject(s)
Endometrium/immunology , Epithelial Cells/immunology , Herpesvirus 6, Human , Infertility/immunology , Roseolovirus Infections/immunology , Trophoblasts/physiology , Adult , Cell Movement , DNA, Viral , Endometrium/virology , Epithelial Cells/virology , Female , Humans , Infertility/virology , Killer Cells, Natural/immunology , Roseolovirus Infections/virology , Stromal Cells/immunology , Stromal Cells/virology , T-Lymphocytes/immunology , Young Adult
9.
Med Microbiol Immunol ; 208(3-4): 475-485, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31065796

ABSTRACT

Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, yet there are no established treatments for preventing maternal-fetal transmission. During first trimester, HCMV replicates in basal decidua that functions as a reservoir for virus and source of transmission to the attached placenta and fetal hemiallograft but also contains immune cells, including natural killer cells, macrophages, and T cell subsets, that respond to pathogens, protecting the placenta and fetus. However, the specific cellular and cytokine responses to infection are unknown, nor are the immune correlates of protection that guide development of therapeutic strategies. Here we survey immune cell phenotypes in intact explants of basal decidua infected with a clinical pathogenic HCMV strain ex vivo and identify specific changes occurring in response to infection in the tissue environment. Using 4-color immunofluorescence microscopy, we found that at 3 days postinfection, virus replicates in decidual stromal cells and epithelial cells of endometrial glands. Infected cells and effector memory CD8+ T cells (TEM) in contact with them make IFN-γ. CD8+ TEM cells produce granulysin and cluster at sites of infection in decidua and the epithelium of endometrial glands. Quantification indicated expansion of two immune cell subtypes-CD8+ TEM cells and, to a lesser extent, iNKT cells. Approximately 20% of immune cells were found in pairs in both control and infected decidua, suggesting frequent cross-talk in the microenvironment of decidua. Our findings indicate a complex immune microenvironment in basal decidua and suggest CD8+ TEM cells play a role in early responses to decidual infection in seropositive women.


Subject(s)
Cytomegalovirus Infections/pathology , Cytomegalovirus/growth & development , Cytomegalovirus/immunology , Decidua/pathology , Immunity, Cellular , Placenta/pathology , CD8-Positive T-Lymphocytes/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Humans , Natural Killer T-Cells/immunology , Organ Culture Techniques , Pregnancy , Stromal Cells/pathology , Stromal Cells/virology
10.
Antiviral Res ; 161: 154-162, 2019 01.
Article in English | MEDLINE | ID: mdl-30481525

ABSTRACT

Herpes simplex virus type-1 (HSV-1) is a significant pathogen that affects vision by targeting multiple regions in the human eye including iris. Using a focused library of synthetic non-saccharide glycosaminoglycan mimetics (NSGMs), we identified sulfated pentagalloylglucoside (SPGG) as a potent inhibitor of HSV-1 entry and cell-to-cell spread in the primary cultures of human iris stromal (HIS) cells isolated from eye donors. Using in vitro ß-galactosidase reporter assay and plaque reduction assay, SPGG was found to inhibit HSV-1 entry in a dosage-dependent manner (IC50 ∼6.0 µM). Interestingly, a pronounced inhibition in HSV-1 entry and spread was observed in HIS cells, or a cell line expressing specific gD-receptor, when virions were pre-treated with mimetics suggesting a possible interaction between SPGG and the HSV-1 glycoprotein. To examine the significance of gD-SPGG interaction, HIS cells were pretreated with SPGG, which showed a significant reduction in gD binding. Taken together, our results provide strong evidence of SPGG being a novel viral entry inhibitor against ocular HSV infection.


Subject(s)
Glucosides/pharmacology , Glycosaminoglycans/pharmacology , Herpesvirus 1, Human/drug effects , Iris/drug effects , Sulfuric Acid Esters/pharmacology , Virus Internalization/drug effects , Cells, Cultured , Glycosaminoglycans/chemical synthesis , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Iris/cytology , Iris/virology , Keratitis, Herpetic/drug therapy , Keratitis, Herpetic/virology , Small Molecule Libraries , Stromal Cells/drug effects , Stromal Cells/virology , Structure-Activity Relationship
11.
Nat Commun ; 9(1): 2436, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29934525

ABSTRACT

Fibroblastic reticular cells (FRCs) are stromal cells in secondary lymphoid organs, the major sites for HIV-1 infection of CD4+ T cells. Although FRCs regulate T cell survival, proliferation, and migration, whether they play any role in HIV-1 spread has not been studied. Here, we show that FRCs enhance HIV-1 spread via trans-infection in which FRCs capture HIV-1 and facilitate infection of T cells that come into contact with FRCs. FRCs mediate trans-infection in both two- and three-dimensional culture systems and in a manner dependent on the virus producer cells. This producer cell dependence, which was also observed for virus spread in secondary lymphoid tissues ex vivo, is accounted for by CD44 incorporated into virus particles and hyaluronan bound to such CD44 molecules. This virus-associated hyaluronan interacts with CD44 expressed on FRCs, thereby promoting virus capture by FRCs. Overall, our results reveal a novel role for FRCs in promoting HIV-1 spread.


Subject(s)
Fibroblasts/metabolism , HIV-1/physiology , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Lymphatic System/metabolism , Blood Buffy Coat , Dendritic Cells , Fibroblasts/virology , HIV-1/pathogenicity , HeLa Cells , Humans , Lymphatic System/virology , Palatine Tonsil , Protein Binding , Stromal Cells/metabolism , Stromal Cells/virology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Tissue Culture Techniques , Virus Replication
12.
Cell Rep ; 23(6): 1794-1805, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29742434

ABSTRACT

During sexual intercourse, HIV-1 crosses epithelial barriers composing the genital mucosa, a poorly understood feature that requires an HIV-1-infected cell vectoring efficient mucosal HIV-1 entry. Therefore, urethral mucosa comprising a polarized epithelium and a stroma composed of fibroblasts and macrophages were reconstructed in vitro. Using this system, we demonstrate by live imaging that efficient HIV-1 transmission to stromal macrophages depends on cell-mediated transfer of the virus through virological synapses formed between HIV-1-infected CD4+ T cells and the epithelial cell mucosal surface. We visualized HIV-1 translocation through mucosal epithelial cells via transcytosis in regions where virological synapses occurred. In turn, interleukin-13 is secreted and HIV-1 targets macrophages, which develop a latent state of infection reversed by lipopolysaccharide (LPS) activation. The live observation of virological synapse formation reported herein is key in the design of vaccines and antiretroviral therapies aimed at blocking HIV-1 access to cellular reservoirs in genital mucosa.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV-1/physiology , Imaging, Three-Dimensional , Immunological Synapses/virology , Macrophages/virology , Adult , Epithelium/virology , HIV Infections/immunology , HIV Infections/virology , Humans , Immunological Synapses/metabolism , Macrophages/pathology , Macrophages/ultrastructure , Male , Models, Biological , Mucous Membrane/virology , Stromal Cells/pathology , Stromal Cells/ultrastructure , Stromal Cells/virology , Urethra/pathology , Virion/metabolism , Virion/ultrastructure
13.
Mol Cancer Res ; 15(10): 1410-1420, 2017 10.
Article in English | MEDLINE | ID: mdl-28679779

ABSTRACT

The tumor microenvironment (TME) is a relevant target for novel biological therapies. MV-m-uPA and MV-h-uPA are fully retargeted, species-specific, oncolytic measles viruses (MV) directed against murine or human urokinase receptor (PLAUR/uPAR), expressed in tumor and stromal cells. The effects of stromal-selective targeting by uPAR-retargeted MVs were investigated. In vitro infection, virus-induced GFP expression, and cytotoxicity by MV-h-uPA and MV-m-uPA were demonstrated in human and murine cancer cells and cancer-associated fibroblasts in a species-specific manner. In a murine fibroblast/human breast cancer 3D coculture model, selective fibroblast targeting by MV-m-uPA inhibited breast cancer cell growth. Systemic administration of murine-specific MV-m-uPA in mice bearing human MDA-MB-231 xenografts was associated with a significant delay in tumor progression and improved survival compared with controls. Experiments comparing tumor (MV-h-uPA) versus stromal (MV-m-uPA) versus combined virus targeting showed that tumor and stromal targeting was associated with improved tumor control over the other groups. Correlative studies confirmed in vivo viral targeting of tumor stroma by MV-m-uPA, increased apoptosis, and virus-induced differential regulation of murine stromal genes associated with inflammatory, angiogenesis, and survival pathways, as well as indirect regulation of human cancer pathways, indicating viral-induced modulation of tumor-stroma interactions. These data demonstrate the feasibility of stromal-selective targeting by an oncolytic MV, virus-induced modulation of tumor-stroma pathways, and subsequent tumor growth delay. These findings further validate the critical role of stromal uPAR in cancer progression and the potential of oncolytic viruses as antistromal agents.Implications: The current report demonstrates for the first time the biological, in vitro, and in vivo antitumor and molecular effects of stromal selective targeting by an oncolytic virus. Mol Cancer Res; 15(10); 1410-20. ©2017 AACR.


Subject(s)
Breast Neoplasms/therapy , Measles virus/physiology , Oncolytic Viruses/physiology , Receptors, Urokinase Plasminogen Activator/genetics , Stromal Cells/cytology , Animals , Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/virology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Female , HT29 Cells , Humans , Mice , Oncolytic Virotherapy , Stromal Cells/virology , Tumor Microenvironment
14.
J Virol ; 91(13)2017 07 01.
Article in English | MEDLINE | ID: mdl-28424285

ABSTRACT

Equine arteritis virus (EAV) has a global impact on the equine industry as the causative agent of equine viral arteritis (EVA), a respiratory, systemic, and reproductive disease of equids. A distinctive feature of EAV infection is that it establishes long-term persistent infection in 10 to 70% of infected stallions (carriers). In these stallions, EAV is detectable only in the reproductive tract, and viral persistence occurs despite the presence of high serum neutralizing antibody titers. Carrier stallions constitute the natural reservoir of the virus as they continuously shed EAV in their semen. Although the accessory sex glands have been implicated as the primary sites of EAV persistence, the viral host cell tropism and whether viral replication in carrier stallions occurs in the presence or absence of host inflammatory responses remain unknown. In this study, dual immunohistochemical and immunofluorescence techniques were employed to unequivocally demonstrate that the ampulla is the main EAV tissue reservoir rather than immunologically privileged tissues (i.e., testes). Furthermore, we demonstrate that EAV has specific tropism for stromal cells (fibrocytes and possibly tissue macrophages) and CD8+ T and CD21+ B lymphocytes but not glandular epithelium. Persistent EAV infection is associated with moderate, multifocal lymphoplasmacytic ampullitis comprising clusters of B (CD21+) lymphocytes and significant infiltration of T (CD3+, CD4+, CD8+, and CD25+) lymphocytes, tissue macrophages, and dendritic cells (Iba-1+ and CD83+), with a small number of tissue macrophages expressing CD163 and CD204 scavenger receptors. This study suggests that EAV employs complex immune evasion mechanisms that warrant further investigation.IMPORTANCE The major challenge for the worldwide control of EAV is that this virus has the distinctive ability to establish persistent infection in the stallion's reproductive tract as a mechanism to ensure its maintenance in equid populations. Therefore, the precise identification of tissue and cellular tropism of EAV is critical for understanding the molecular basis of viral persistence and for development of improved prophylactic or treatment strategies. This study significantly enhances our understanding of the EAV carrier state in stallions by unequivocally identifying the ampullae as the primary sites of viral persistence, combined with the fact that persistence involves continuous viral replication in fibrocytes (possibly including tissue macrophages) and T and B lymphocytes in the presence of detectable inflammatory responses, suggesting the involvement of complex viral mechanisms of immune evasion. Therefore, EAV persistence provides a powerful new natural animal model to study RNA virus persistence in the male reproductive tract.


Subject(s)
B-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Epithelium/virology , Equartevirus/physiology , Genitalia/virology , Stromal Cells/virology , Viral Tropism , Animals , Arterivirus Infections/veterinary , Arterivirus Infections/virology , Fluorescent Antibody Technique , Horse Diseases/virology , Horses , Immunohistochemistry , Male
15.
Sci Rep ; 7: 44286, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281680

ABSTRACT

Zika virus (ZIKV) is a recently re-emerged flavivirus transmitted to humans by mosquito bites but also from mother to fetus and by sexual intercourse. We here show that primary human endometrial stromal cells (HESC) are highly permissive to ZIKV infection and support its in vitro replication. ZIKV envelope expression was detected in the endoplasmic reticulum whereas double-stranded viral RNA colocalized with vimentin filaments to the perinuclear region. ZIKV productive infection also occurred in the human T-HESC cell line together with the induction of interferon-ß (IFN-ß) and of IFN-stimulated genes. Notably, in vitro decidualization of T-HESC with cyclic AMP and progesterone upregulated the cell surface expression of the ZIKV entry co-receptor AXL and boosted ZIKV replication by ca. 100-fold. Thus, endometrial stromal cells, particularly if decidualized, likely represent a crucial cell target of ZIKV reaching them, either via the uterine vasculature in the viremic phase of the infection or by sexual viral transmission, and a potential source of virus spreading to placental trophoblasts during pregnancy.


Subject(s)
Endometrium/virology , Stromal Cells/virology , Virus Replication/physiology , Zika Virus/physiology , Adult , Animals , Cell Line , Cells, Cultured , Chlorocebus aethiops , Endometrium/cytology , Female , Gene Expression , Host-Pathogen Interactions/genetics , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Replication/genetics , Zika Virus/genetics , Zika Virus Infection/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/virology
16.
J Infect Dis ; 215(1): 64-69, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27226206

ABSTRACT

Here we describe clinicopathologic features of Ebola virus disease in pregnancy. One woman infected with Sudan virus in Gulu, Uganda, in 2000 had a stillbirth and survived, and another woman infected with Bundibugyo virus had a live birth with maternal and infant death in Isiro, the Democratic Republic of the Congo in 2012. Ebolavirus antigen was seen in the syncytiotrophoblast and placental maternal mononuclear cells by immunohistochemical analysis, and no antigen was seen in fetal placental stromal cells or fetal organs. In the Gulu case, ebolavirus antigen localized to malarial parasite pigment-laden macrophages. These data suggest that trophoblast infection may be a mechanism of transplacental ebolavirus transmission.


Subject(s)
Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Pregnancy Complications, Infectious/pathology , Pregnancy Complications, Infectious/virology , Adult , Antibodies, Viral/blood , Antigens, Viral/immunology , Antigens, Viral/isolation & purification , Democratic Republic of the Congo , Ebolavirus/chemistry , Ebolavirus/genetics , Ebolavirus/immunology , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/transmission , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunohistochemistry , Macrophages/parasitology , Macrophages/ultrastructure , Macrophages/virology , Malaria/complications , Malaria/immunology , Malaria/virology , Microscopy, Electron, Transmission , Placenta/ultrastructure , Placenta/virology , Polymerase Chain Reaction , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/parasitology , Stillbirth , Stromal Cells/ultrastructure , Stromal Cells/virology , Trophoblasts/parasitology , Trophoblasts/ultrastructure , Trophoblasts/virology
17.
Prog Mol Biol Transl Sci ; 144: 169-238, 2016.
Article in English | MEDLINE | ID: mdl-27865458

ABSTRACT

Human papillomaviruses (HPVs) are small, double-stranded DNA viruses that replicate in stratified squamous epithelia and cause a variety of malignancies. Current efforts in HPV biology are focused on understanding the virus-host interactions that enable HPV to persist for years or decades in the tissue. The importance of interactions between tumor cells and the stromal microenvironment has become increasingly apparent in recent years, but how stromal interactions impact the normal, benign life cycle of HPVs, or progression of lesions to cancer is less understood. Furthermore, how productively replicating HPV impacts cells in the stromal environment is also unclear. Here we bring together some of the relevant literature on keratinocyte-stromal interactions and their impacts on HPV biology, focusing on stromal fibroblasts, immune cells, and endothelial cells. We discuss how HPV oncogenes in infected cells manipulate other cells in their environment, and, conversely, how neighboring cells may impact the efficiency or course of HPV infection.


Subject(s)
Cellular Microenvironment , Papillomaviridae/physiology , Animals , Extracellular Matrix/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Stromal Cells/virology
18.
Biol Reprod ; 95(1): 12, 2016 07.
Article in English | MEDLINE | ID: mdl-27281703

ABSTRACT

Viral infections can cause genital tract disorders (including abortion) in cows, and bovine herpesvirus 4 (BoHV-4) is often present in endometritis-affected animals. A major problem with cattle uterine viral infections in general, and BoHV-4 in particular, is our limited understanding of the pathogenic role(s) that these infections play in the endometrium. A similar lack of knowledge holds for the molecular mechanisms utilized, and the host cell pathways affected, by BoHV-4. To begin to fill these gaps, we set up optimized conditions for BoHV-4 infection of a pure population of bovine endometrial stromal cells (BESCs) to be used as source material for RNA sequencing-based transcriptome profiling. Many genes were found to be upregulated (417) or downregulated (181) after BoHV-4 infection. As revealed by enrichment functional analysis on differentially expressed genes, BoHV-4 infection affects various pathways related to cell proliferation and cell surface integrity, at least three of which were centered on upregulation of matrix metalloproteinase 1 (MMP1) and interleukin 8 (IL8). This was confirmed by reverse transcription PCR, real-time PCR, Western-immunoblot analysis, and a luciferase assay with a bovine MMP1-specific promoter reporter construct. Further, it was found that MMP1 transcription was upregulated by the BoHV-4 transactivator IE2/RTA, leading to abnormally high metalloproteinase tissue levels, potentially leading to defective endometrium healing and unresolved inflammation. Based on these findings, we propose a new model for BoHV-4 action centered on IE2-mediated MMP1 upregulation and novel therapeutic interventions based on IFN gamma-mediated MMP1 downregulation.


Subject(s)
Endometrium/metabolism , Herpesvirus 4, Bovine , Matrix Metalloproteinase 1/metabolism , Stromal Cells/metabolism , Up-Regulation , Animals , Cattle , Endometrium/pathology , Endometrium/virology , Female , Gene Expression Profiling , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Interleukin-8/genetics , Interleukin-8/metabolism , Matrix Metalloproteinase 1/genetics , Stromal Cells/pathology , Stromal Cells/virology
19.
Immunity ; 44(3): 622-633, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26921107

ABSTRACT

Stromal cells generate a complex cellular scaffold that provides specialized microenvironments for lymphocyte activation in secondary lymphoid organs. Here, we assessed whether local activation of stromal cells in the central nervous system (CNS) is mandatory to transfer immune recognition from secondary lymphoid organs into the infected tissue. We report that neurotropic virus infection in mice triggered the establishment of such stromal cell niches in the CNS. CNS stromal cell activation was dominated by a rapid and vigorous production of CC-motif chemokine receptor (CCR) 7 ligands CCL19 and CCL21 by vascular endothelial cells and adjacent fibroblastic reticular cell (FRC)-like cells in the perivascular space. Moreover, CCR7 ligands produced by CNS stromal cells were crucial to support recruitment and local re-activation of antiviral CD8(+) T cells and to protect the host from lethal neuroinflammatory disease, indicating that CNS stromal cells generate confined microenvironments that control protective T cell immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Endothelium, Vascular/immunology , Hepatitis A virus/immunology , Hepatitis A/immunology , Neurogenic Inflammation/parasitology , Receptors, CCR7/metabolism , Stromal Cells/immunology , Animals , Cell Movement , Cellular Microenvironment , Central Nervous System/virology , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Endothelium, Vascular/virology , Hepatitis A/complications , Immunity, Cellular , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurogenic Inflammation/etiology , Receptors, CCR7/genetics , Stromal Cells/virology , Viral Tropism
20.
PLoS Pathog ; 11(9): e1005153, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26402858

ABSTRACT

Two of the crucial aspects of human immunodeficiency virus (HIV) infection are (i) viral persistence in reservoirs (precluding viral eradication) and (ii) chronic inflammation (directly associated with all-cause morbidities in antiretroviral therapy (ART)-controlled HIV-infected patients). The objective of the present study was to assess the potential involvement of adipose tissue in these two aspects. Adipose tissue is composed of adipocytes and the stromal vascular fraction (SVF); the latter comprises immune cells such as CD4+ T cells and macrophages (both of which are important target cells for HIV). The inflammatory potential of adipose tissue has been extensively described in the context of obesity. During HIV infection, the inflammatory profile of adipose tissue has been revealed by the occurrence of lipodystrophies (primarily related to ART). Data on the impact of HIV on the SVF (especially in individuals not receiving ART) are scarce. We first analyzed the impact of simian immunodeficiency virus (SIV) infection on abdominal subcutaneous and visceral adipose tissues in SIVmac251 infected macaques and found that both adipocytes and adipose tissue immune cells were affected. The adipocyte density was elevated, and adipose tissue immune cells presented enhanced immune activation and/or inflammatory profiles. We detected cell-associated SIV DNA and RNA in the SVF and in sorted CD4+ T cells and macrophages from adipose tissue. We demonstrated that SVF cells (including CD4+ T cells) are infected in ART-controlled HIV-infected patients. Importantly, the production of HIV RNA was detected by in situ hybridization, and after the in vitro reactivation of sorted CD4+ T cells from adipose tissue. We thus identified adipose tissue as a crucial cofactor in both viral persistence and chronic immune activation/inflammation during HIV infection. These observations open up new therapeutic strategies for limiting the size of the viral reservoir and decreasing low-grade chronic inflammation via the modulation of adipose tissue-related pathways.


Subject(s)
Adipose Tissue/virology , Disease Reservoirs , HIV Infections/virology , HIV/physiology , Panniculitis/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Coculture Techniques , Female , HIV/immunology , HIV/isolation & purification , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/pathology , Host-Pathogen Interactions , Humans , Immunity, Innate , Macaca fascicularis , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Macrophages/virology , Male , Middle Aged , Panniculitis/immunology , Panniculitis/metabolism , Panniculitis/pathology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/isolation & purification , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Stromal Cells/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...