Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 664
Filter
1.
Methods Mol Biol ; 2807: 299-323, 2024.
Article in English | MEDLINE | ID: mdl-38743237

ABSTRACT

Ex vivo cervical tissue explant models offer a physiologically relevant approach for studying virus-host interactions that underlie mucosal HIV-1 transmission to women. However, the utility of cervical explant tissue (CET) models has been limited for both practical and technical reasons. These include assay variation, inadequate sensitivity for assessing HIV-1 infection and replication in tissue, and constraints imposed by the requirement for using multiple replica samples of CET to test each experimental variable and assay parameter. Here, we describe an experimental approach that employs secreted nanoluciferase (sNLuc) and current HIV-1 reporter virus technologies to overcome certain limitations of earlier ex vivo CET models. This method augments application of the CET model for investigating important questions involving mucosal HIV-1 transmission.


Subject(s)
Cervix Uteri , HIV Infections , HIV-1 , HIV-1/physiology , HIV-1/genetics , Humans , Cervix Uteri/virology , Cervix Uteri/metabolism , Female , HIV Infections/virology , Luciferases/genetics , Luciferases/metabolism , Genes, Reporter , Mucous Membrane/virology , Mucous Membrane/metabolism , Virus Replication
2.
Nat Microbiol ; 9(5): 1231-1243, 2024 May.
Article in English | MEDLINE | ID: mdl-38649413

ABSTRACT

The 2022 mpox virus (MPXV) outbreak was sustained by human-to-human transmission; however, it is currently unclear which factors lead to sustained transmission of MPXV. Here we present Mastomys natalensis as a model for MPXV transmission after intraperitoneal, rectal, vaginal, aerosol and transdermal inoculation with an early 2022 human outbreak isolate (Clade IIb). Virus shedding and tissue replication were route dependent and occurred in the presence of self-resolving localized skin, lung, reproductive tract or rectal lesions. Mucosal inoculation via the rectal, vaginal and aerosol routes led to increased shedding, replication and a pro-inflammatory T cell profile compared with skin inoculation. Contact transmission was higher from rectally inoculated animals. This suggests that transmission might be sustained by increased susceptibility of the anal and genital mucosae for infection and subsequent virus release.


Subject(s)
Mucous Membrane , Poxviridae Infections , Virus Shedding , Animals , Female , Mucous Membrane/virology , Poxviridae Infections/transmission , Poxviridae Infections/virology , Poxviridae Infections/veterinary , Humans , Virus Replication , Disease Models, Animal , Rodentia/virology , Male , Rats , Vagina/virology , Disease Outbreaks
3.
Proc Natl Acad Sci U S A ; 119(27): e2116197119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35767643

ABSTRACT

The majority of viruses within the gut are obligate bacterial viruses known as bacteriophages (phages). Their bacteriotropism underscores the study of phage ecology in the gut, where they modulate and coevolve with gut bacterial communities. Traditionally, these ecological and evolutionary questions were investigated empirically via in vitro experimental evolution and, more recently, in vivo models were adopted to account for physiologically relevant conditions of the gut. Here, we probed beyond conventional phage-bacteria coevolution to investigate potential tripartite evolutionary interactions between phages, their bacterial hosts, and the mammalian gut mucosa. To capture the role of the mammalian gut, we recapitulated a life-like gut mucosal layer using in vitro lab-on-a-chip devices (to wit, the gut-on-a-chip) and showed that the mucosal environment supports stable phage-bacteria coexistence. Next, we experimentally coevolved lytic phage populations within the gut-on-a-chip devices alongside their bacterial hosts. We found that while phages adapt to the mucosal environment via de novo mutations, genetic recombination was the key evolutionary force in driving mutational fitness. A single mutation in the phage capsid protein Hoc-known to facilitate phage adherence to mucus-caused altered phage binding to fucosylated mucin glycans. We demonstrated that the altered glycan-binding phenotype provided the evolved mutant phage a competitive fitness advantage over its ancestral wild-type phage in the gut-on-a-chip mucosal environment. Collectively, our findings revealed that phages-in addition to their evolutionary relationship with bacteria-are able to evolve in response to a mammalian-derived mucosal environment.


Subject(s)
Bacteria , Bacteriophages , Gastrointestinal Tract , Mucous Membrane , Animals , Bacteria/virology , Bacteriophages/genetics , Bacteriophages/physiology , Capsid Proteins/genetics , Gastrointestinal Tract/virology , Mucous Membrane/virology , Mucus , Mutation , Symbiosis
4.
J Virol ; 96(10): e0187521, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35475668

ABSTRACT

Persistent infection with some mucosal α-genus human papillomaviruses (HPVs; the most prevalent one being HPV16) can induce cervical carcinoma, anogenital cancers, and a subset of head and neck squamous cell carcinoma (HNSCC). Cutaneous ß-genus HPVs (such as HPV5 and HPV8) associate with skin lesions that can progress into squamous cell carcinoma with sun exposure in Epidermodysplasia verruciformis patients and immunosuppressed patients. Here, we analyzed mechanisms used by E6 proteins from the α- and ß-genus to inhibit the interferon-ß (IFNB1) response. HPV16 E6 mediates this effect by a strong direct interaction with interferon regulatory factor 3 (IRF3). The binding site of E6 was localized within a flexible linker between the DNA-binding domain and the IRF-activation domain of IRF3 containing an LxxLL motif. The crystallographic structure of the complex between HPV16 E6 and the LxxLL motif of IRF3 was solved and compared with the structure of HPV16 E6 interacting with the LxxLL motif of the ubiquitin ligase E6AP. In contrast, cutaneous HPV5 and HPV8 E6 proteins bind to the IRF3-binding domain (IBiD) of the CREB-binding protein (CBP), a key transcriptional coactivator in IRF3-mediated IFN-ß expression. IMPORTANCE Persistent HPV infections can be associated with the development of several cancers. The ability to persist depends on the ability of the virus to escape the host immune system. The type I interferon (IFN) system is the first-line antiviral defense strategy. HPVs carry early proteins that can block the activation of IFN-I. Among mucosal α-genus HPV types, the HPV16 E6 protein has a remarkable property to strongly interact with the transcription factor IRF3. Instead, cutaneous HPV5 and HPV8 E6 proteins bind to the IRF3 cofactor CBP. These results highlight the versatility of E6 proteins to interact with different cellular targets. The interaction between the HPV16 E6 protein and IRF3 might contribute to the higher prevalence of HPV16 than that of other high-risk mucosal HPV types in HPV-associated cancers.


Subject(s)
Interferon Regulatory Factor-3 , Interferon-beta , Oncogene Proteins, Viral , Papillomavirus Infections , Repressor Proteins , Human papillomavirus 16/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mucous Membrane/virology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Skin/virology
5.
Viruses ; 14(2)2022 02 15.
Article in English | MEDLINE | ID: mdl-35215986

ABSTRACT

Infectious bursal disease virus (IBDV) is one of the most important infectious diseases of poultry around the world. Gut-associated lymphoid tissues (GALT) are the first line of defense of the host against the infection. The purpose of this study was to investigate the role of innate immune antiviral signaling triggered by Toll-like receptor 3 (TLR3), as well as macrophage activation and cytokine response in the intestinal lamina propria (ILP) cells after the oral challenge of IBDV in relation to IBDV virulence and disease pathogenesis. The results showed that the expression levels of TLR3, IRF7, IFN-α/ß and the corresponding downstream antiviral factors OAS, PKR and Mx were all upregulated in the SPF chicken ILP cells at 8 h post-infection (hpi) and 12 hpi. Similarly, macrophages were activated, with the initial macrophage M1 activation observed at 8 hpi, but then it rapidly shifted to a non-protective M2-type. Both Th1 (IFN-γ, TNF-α, IL-12) and Th2 (IL-4 and IL-10) types of cytokines were differentially upregulated during the early stage of infection; however, the Th1 cytokines exhibited stronger activation before 8 hpi compared to those of the Th2 cytokines. Interestingly, differential regulations of gene expression induced by different IBDV strains with different virulence were detected. The HLJ0504-like very virulent (vv) IBDV strain NN1172 induced stronger activation of TLR3-IFN-α/ß pathway, macrophages and the Th1/2 cytokines' expression, compared to those induced by the attenuated strain B87 at 8 hpi and 12 hpi in the ILP cells. In conclusion, the innate antiviral response mediated by the TLR3-IRF7 pathway, macrophage activation and cytokine expression in the GALT cells at the early stage of IBDV infection was differentially modulated, and the HLJ0504-like vvIBDV strain triggered stronger activation than the attenuated vaccine strain, and that may play an important role in the progression of disease.


Subject(s)
Birnaviridae Infections/veterinary , Infectious bursal disease virus/pathogenicity , Mucous Membrane/virology , Poultry Diseases/pathology , Animals , Birnaviridae Infections/pathology , Birnaviridae Infections/virology , Chick Embryo , Chickens/virology , Cytokines/metabolism , Immunity, Innate , Mucous Membrane/pathology , Poultry Diseases/virology , Virulence
6.
PLoS One ; 17(1): e0262108, 2022.
Article in English | MEDLINE | ID: mdl-35030192

ABSTRACT

Viruses are common components of the intestinal microbiome, modulating host bacterial metabolism and interacting with the immune system, with a possible role in the pathogenesis of immune-mediated diseases such as celiac disease (CeD). The objective of this study was to characterize the virome profile in children with new-onset CeD. We used metagenomic analysis of viral DNA in mucosal and fecal samples from children with CeD and controls and performed sequencing using the Nextera XT library preparation kit. Abundance log2 fold changes were calculated using differential expression and linear discriminant effect size. Shannon alpha and Bray-Curtis beta diversity were determined. A total of 40 children with CeD and 39 controls were included. We found viral dysbiosis in both fecal and mucosal samples. Examples of significantly more abundant species in fecal samples of children with CeD included Human polyomavirus 2, Enterobacteria phage mEpX1, and Enterobacteria phage mEpX2; whereas less abundant species included Lactococcus phages ul36 and Streptococcus phage Abc2. In mucosal samples however, no species were significantly associated with CeD. Shannon alpha diversity was not significantly different between CeD and non-CeD groups and Bray-Curtis beta diversity showed no significant separation between CeD and non-CeD samples in either mucosal or stool samples, whereas separation was clear in all samples. We identified significant viral dysbiosis in children with CeD, suggesting a potential role in the pathogenesis of CeD indicating the need for further studies.


Subject(s)
Celiac Disease/virology , Dysbiosis/diagnosis , Metagenomics/methods , Sequence Analysis, DNA/methods , Viruses/classification , Adolescent , Age of Onset , Case-Control Studies , Child , DNA, Viral/genetics , Dysbiosis/virology , Feces/virology , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Mucous Membrane/virology , Phylogeny , Viruses/genetics , Viruses/isolation & purification
7.
Sci Rep ; 11(1): 23514, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34873252

ABSTRACT

Inflammatory cytokines augment humoral responses by stimulating antibody production and inducing class-switching. In women, genital inflammation (GI) significantly modifies HIV risk. However, the impact of GI on mucosal antibodies remains undefined. We investigated the impact of GI, pre-HIV infection, on antibody isotypes and IgG subclasses in the female genital tract. Immunoglobulin (Ig) isotypes, IgG subclasses and 48 cytokines were measured prior to HIV infection in cervicovaginal lavages (CVL) from 66 HIV seroconverters (cases) and 66 matched HIV-uninfected women (controls) enrolled in the CAPRISA 004 and 008 1% tenofovir gel trials. Pre-HIV infection, cases had significantly higher genital IgM (4.13; IQR, 4.04-4.19) compared to controls (4.06; IQR, 3.90-4.20; p = 0.042). More than one-quarter of cases (27%) had GI compared to just over one-tenth (12%) in controls. Significantly higher IgG1, IgG3, IgG4 and IgM (all p < 0.05) were found in women stratified for GI compared to women without. Adjusted linear mixed models showed several pro-inflammatory, chemotactic, growth factors, and adaptive cytokines significantly correlated with higher titers of IgM, IgA and IgG subclasses (p < 0.05). The strong and significant positive correlations between mucosal antibodies and markers of GI suggest that GI may impact mucosal antibody profiles. These findings require further investigation to establish a plausible biological link between the local inflammatory milieu and its consequence on these genital antibodies.


Subject(s)
Antibodies/immunology , Genitalia, Female/immunology , HIV Antibodies/immunology , Inflammation/immunology , Mucous Membrane/immunology , Adolescent , Case-Control Studies , Cytokines/immunology , Double-Blind Method , Female , Genitalia, Female/virology , Humans , Immunoglobulins/immunology , Inflammation/virology , Mucous Membrane/virology , Retrospective Studies , Tenofovir/immunology
8.
Front Immunol ; 12: 785072, 2021.
Article in English | MEDLINE | ID: mdl-34956215

ABSTRACT

Background: The vasodilator neuropeptide calcitonin gene-related peptide (CGRP) plays both detrimental and protective roles in different pathologies. CGRP is also an essential component of the neuro-immune dialogue between nociceptors and mucosal immune cells. We previously discovered that CGRP is endowed with anti-viral activity and strongly inhibits human immunodeficiency virus type 1 (HIV-1) infection, by suppressing Langerhans cells (LCs)-mediated HIV-1 trans-infection in-vitro and mucosal HIV-1 transmission ex-vivo. This inhibition is mediated via activation of the CGRP receptor non-canonical NFκB/STAT4 signaling pathway that induces a variety of cooperative mechanisms. These include CGRP-mediated increase in the expression of the LC-specific pathogen recognition C-type lectin langerin and decrease in LC-T-cell conjugates formation. The clinical utility of CGRP and modalities of CGRP receptor activation, for inhibition of mucosal HIV-1 transmission, remain elusive. Methods: We tested the capacity of CGRP to inhibit HIV-1 infection in-vivo in humanized mice. We further compared the anti-HIV-1 activities of full-length native CGRP, its metabolically stable analogue SAX, and several CGRP peptide fragments containing its binding C-terminal and activating N-terminal regions. These agonists were evaluated for their capacity to inhibit LCs-mediated HIV-1 trans-infection in-vitro and mucosal HIV-1 transmission in human mucosal tissues ex-vivo. Results: A single CGRP intravaginal topical treatment of humanized mice, followed by HIV-1 vaginal challenge, transiently restricts the increase in HIV-1 plasma viral loads but maintains long-lasting higher CD4+ T-cell counts. Similarly to CGRP, SAX inhibits LCs-mediated HIV-1 trans-infection in-vitro, but with lower potency. This inhibition is mediated via CGRP receptor activation, leading to increased expression of both langerin and STAT4 in LCs. In contrast, several N-terminal and N+C-terminal bivalent CGRP peptide fragments fail to increase langerin and STAT4, and accordingly lack anti-HIV-1 activities. Finally, like CGRP, treatment of human inner foreskin tissue explants with SAX, followed by polarized inoculation with cell-associated HIV-1, completely blocks formation of LC-T-cell conjugates and HIV-1 infection of T-cells. Conclusion: Our results show that CGRP receptor activation by full-length CGRP or SAX is required for efficient inhibition of LCs-mediated mucosal HIV-1 transmission. These findings suggest that formulations containing CGRP, SAX and/or their optimized agonists/analogues could be harnessed for HIV-1 prevention.


Subject(s)
Calcitonin Gene-Related Peptide/pharmacology , HIV Infections/prevention & control , Peptide Fragments/pharmacology , T-Lymphocytes/drug effects , Animals , Calcitonin Gene-Related Peptide/therapeutic use , Dipeptides/pharmacology , Disease Models, Animal , Female , HEK293 Cells , HIV Infections/diagnosis , HIV Infections/transmission , HIV Infections/virology , HIV-1/isolation & purification , HIV-1/pathogenicity , Healthy Volunteers , Humans , Mice , Mucous Membrane/drug effects , Mucous Membrane/immunology , Mucous Membrane/virology , Peptide Fragments/therapeutic use , Primary Cell Culture , Quinazolines/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tissue Culture Techniques
9.
Front Immunol ; 12: 778455, 2021.
Article in English | MEDLINE | ID: mdl-34868050

ABSTRACT

Introduction: Acetylsalicylic acid (ASA) is a well-known and safe anti-inflammatory. At low-dose, it is prescribed to prevent secondary cardiovascular events in those with pre-existing conditions and to prevent preeclampsia. Little is known about how low-dose ASA affects the immune response. In this study, we followed women to assess how ASA use modifies T cells immune phenotypes in the blood and at the genital tract. Methods: HIV uninfected women from Kenya were enrolled in this study and followed for one month to assess baseline responses including systemic/mucosal baseline immune activation. Participants then received 81mg of ASA daily for 6 weeks to assess changes to T cell immune activation (systemic and mucosal) relative to baseline levels. Results: The concentration of ASA measured in the blood was 58% higher than the level measured at the female genital tract. In the blood, the level of ASA was inversely correlated with the following: the proportion of Th17 expressing HLA-DR (p=0.04), the proportion of effector CD4+ T cells expressing CCR5 (p=0.03) and the proportion of CD8+Tc17 expressing CCR5 (p=0.04). At the genital tract, ASA use correlated with a decreased of activated CD4+T cells [CD4+CCR5+CD161+ (p=0.02) and CD4+CCR5+CD95+ (p=0.001)]. Conclusion: This study shows that ASA use impacts the immune response in both the systemic and genital tract compartments. This could have major implications for the prevention of infectious diseases such as HIV, in which the virus targets activated T cells to establish an infection. This could inform guidelines on ASA use in women. Clinical Trial Registration: ClinicalTrials.gov, identifier NCT02079077.


Subject(s)
Aspirin/administration & dosage , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Adult , Anti-Inflammatory Agents/pharmacology , Biomarkers , Cytokines/metabolism , Female , HIV Infections/epidemiology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , Humans , Immunity, Mucosal , Kenya/epidemiology , Leukocytes, Mononuclear , Male , Middle Aged , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/virology , T-Lymphocytes/metabolism , Young Adult
10.
PLoS One ; 16(12): e0260010, 2021.
Article in English | MEDLINE | ID: mdl-34855793

ABSTRACT

Little is known about how specific individual viral lineages replicating systemically during acute Human Immunodeficiency Virus or Simian Immunodeficiency Virus (HIV/SIV) infection persist into chronic infection. In this study, we use molecularly barcoded SIV (SIVmac239M) to track distinct viral lineages for 12 weeks after intravenous (IV) or intrarectal (IR) challenge in macaques. Two Mafa-A1*063+ cynomolgus macaques (Macaca fascicularis, CM) were challenged IV, and two Mamu-A1*001+ rhesus macaques (Macaca mulatta, RM) were challenged IR with 200,000 Infectious Units (IU) of SIVmac239M. We sequenced the molecular barcode of SIVmac239M from all animals over the 12 weeks of the study to characterize the diversity and persistence of virus lineages. During the first three weeks post-infection, we found ~70-560 times more unique viral lineages circulating in the animals challenged IV compared to those challenged IR, which is consistent with the hypothesis that the challenge route is the primary driver restricting the transmission of individual viral lineages. We also characterized the sequences of T cell epitopes targeted during acute SIV infection, and found that the emergence of escape variants in acutely targeted epitopes can occur on multiple virus templates simultaneously, but that elimination of some of these templates is likely a consequence of additional host factors. These data imply that virus lineages present during acute infection can still be eliminated from the systemic virus population even after initial selection.


Subject(s)
Mucous Membrane/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/growth & development , Animals , Epitopes/immunology , Female , Gene Products, tat/genetics , Injections, Intravenous , Macaca fascicularis/immunology , Macaca fascicularis/virology , Macaca mulatta/immunology , Macaca mulatta/virology , Mucous Membrane/immunology , Mutation , RNA, Viral/blood , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Load , Viremia/immunology , Viremia/virology
11.
Viruses ; 13(10)2021 10 11.
Article in English | MEDLINE | ID: mdl-34696473

ABSTRACT

Mother-to-child transmission (MTCT) of HIV-1 may occur during pregnancy, labor, and breastfeeding; however, the molecular mechanism of MTCT of virus remains poorly understood. Infant tonsil mucosal epithelium may sequester HIV-1, serving as a transient reservoir, and may play a critical role in MTCT. Innate immune proteins human beta-defensins 2 (hBD-2) and -3 may inactivate intravesicular virions. To establish delivery of hBD-2 and -3 into vesicles containing HIV-1, we tagged hBDs with the protein transduction domain (PTD) of HIV-1 Tat, which facilitates an efficient translocation of proteins across cell membranes. Our new findings showed that hBD-2 and -3 proteins tagged with PTD efficiently penetrated polarized tonsil epithelial cells by endocytosis and direct penetration. PTD-initiated internalization of hBD-2 and -3 proteins into epithelial cells led to their subsequent penetration of multivesicular bodies (MVB) and vacuoles containing HIV-1. Furthermore, PTD played a role in the fusion of vesicles containing HIV-1 with lysosomes, where virus was inactivated. PTD-initiated internalization of hBD-2 and -3 proteins into ex vivo tonsil tissue explants reduced the spread of virus from epithelial cells to CD4+ T lymphocytes, CD68+ macrophages, and CD1c+ dendritic cells, suggesting that this approach may serve as an antiviral strategy for inactivating intraepithelial HIV-1 and reducing viral MTCT.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/virology , HIV-1/physiology , Palatine Tonsil/virology , beta-Defensins/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , CD4-Positive T-Lymphocytes , Endocytosis , Epithelium , HIV Infections , Humans , Infectious Disease Transmission, Vertical , Macrophages/virology , Mucous Membrane/virology , Protein Domains , beta-Defensins/genetics , tat Gene Products, Human Immunodeficiency Virus/chemistry
12.
Front Immunol ; 12: 724618, 2021.
Article in English | MEDLINE | ID: mdl-34484233

ABSTRACT

Herpes simplex virus type 2 (HSV-2) infection is one of the most prevalent sexually transmitted infections that disproportionately impacts women worldwide. Currently, there are no vaccines or curative treatments, resulting in life-long infection. The mucosal environment of the female reproductive tract (FRT) is home to a complex array of local immune defenses that must be carefully coordinated to protect against genital HSV-2 infection, while preventing excessive inflammation to prevent disease symptoms. Crucial to the defense against HSV-2 infection in the FRT are three classes of highly related and integrated cytokines, type I, II, and III interferons (IFN). These three classes of cytokines control HSV-2 infection and reduce tissue damage through a combination of directly inhibiting viral replication, as well as regulating the function of resident immune cells. In this review, we will examine how interferons are induced and their critical role in how they shape the local immune response to HSV-2 infection in the FRT.


Subject(s)
Herpes Genitalis/immunology , Herpesvirus 2, Human/immunology , Interferons/immunology , Animals , Female , Humans , Immunity, Mucosal , Mice , Mucous Membrane/virology
13.
EMBO J ; 40(20): e106765, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34510494

ABSTRACT

The current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and outbreaks of new variants highlight the need for preventive treatments. Here, we identified heparan sulfate proteoglycans as attachment receptors for SARS-CoV-2. Notably, neutralizing antibodies against SARS-CoV-2 isolated from COVID-19 patients interfered with SARS-CoV-2 binding to heparan sulfate proteoglycans, which might be an additional mechanism of antibodies to neutralize infection. SARS-CoV-2 binding to and infection of epithelial cells was blocked by low molecular weight heparins (LMWH). Although dendritic cells (DCs) and mucosal Langerhans cells (LCs) were not infected by SARS-CoV-2, both DC subsets efficiently captured SARS-CoV-2 via heparan sulfate proteoglycans and transmitted the virus to ACE2-positive cells. Notably, human primary nasal cells were infected by SARS-CoV-2, and infection was blocked by pre-treatment with LMWH. These data strongly suggest that heparan sulfate proteoglycans are important attachment receptors facilitating infection and transmission, and support the use of LMWH as prophylaxis against SARS-CoV-2 infection.


Subject(s)
COVID-19/transmission , Heparan Sulfate Proteoglycans/metabolism , Heparin, Low-Molecular-Weight/pharmacology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/pharmacology , Chlorocebus aethiops , Dendritic Cells/metabolism , Dendritic Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Host-Pathogen Interactions , Humans , Mucous Membrane/cytology , Mucous Membrane/virology , SARS-CoV-2/metabolism , Syndecan-1/metabolism , Syndecan-4/metabolism , Vero Cells , COVID-19 Drug Treatment
14.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Article in English | MEDLINE | ID: mdl-34583992

ABSTRACT

Membrane-associated mucins protect epithelial cell surfaces against pathogenic threats by serving as nonproductive decoys that capture infectious agents and clear them from the cell surface and by erecting a physical barrier that restricts their access to target receptors on host cells. However, the mechanisms through which mucins function are still poorly defined because of a limited repertoire of tools available for tailoring their structure and composition in living cells with molecular precision. Using synthetic glycopolymer mimetics of mucins, we modeled the mucosal glycocalyx on red blood cells (RBCs) and evaluated its influence on lectin (SNA) and virus (H1N1) adhesion to endogenous sialic acid receptors. The glycocalyx inhibited the rate of SNA and H1N1 adhesion in a size- and density-dependent manner, consistent with the current view of mucins as providing a protective shield against pathogens. Counterintuitively, increasing the density of the mucin mimetics enhanced the retention of bound lectins and viruses. Careful characterization of SNA behavior at the RBC surface using a range of biophysical and imaging techniques revealed lectin-induced crowding and reorganization of the glycocalyx with concomitant enhancement in lectin clustering, presumably through the formation of a more extensive glycan receptor patch at the cell membrane. Our findings indicate that glycan-targeting pathogens may exploit the biophysical and biomechanical properties of mucins to overcome the mucosal glycocalyx barrier.


Subject(s)
Erythrocytes/metabolism , Glycocalyx/metabolism , Lectins/metabolism , Mucins/metabolism , Polysaccharides/metabolism , Biomimetics/methods , Cell Membrane/metabolism , Cell Membrane/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Erythrocytes/virology , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Mucous Membrane/metabolism , Mucous Membrane/virology , Receptors, Cell Surface/metabolism
15.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34578405

ABSTRACT

Papillomavirus L1 and L2, the major and minor capsid proteins, play significant roles in viral assembly, entry, and propagation. In the current study, we investigate the impact of L1 and L2 on viral life cycle and tumor growth with a newly established mouse papillomavirus (MmuPV1) infection model. MmuPV1 L1 knockout, L2 knockout, and L1 plus L2 knockout mutant genomes (designated as L1ATGko-4m, L2ATGko, and L1-L2ATGko respectively) were generated. The mutants were examined for their ability to generate lesions in athymic nude mice. Viral activities were examined by qPCR, immunohistochemistry (IHC), in situ hybridization (ISH), and transmission electron microscopy (TEM) analyses. We demonstrated that viral DNA replication and tumor growth occurred at both cutaneous and mucosal sites infected with each of the mutants. Infections involving L1ATGko-4m, L2ATGko, and L1-L2ATGko mutant genomes generally resulted in smaller tumor sizes compared to infection with the wild type. The L1 protein was absent in L1ATGko-4m and L1-L2ATGko mutant-treated tissues, even though viral transcripts and E4 protein expression were robust. Therefore, L1 is not essential for MmuPV1-induced tumor growth, and this finding parallels our previous observations in the rabbit papillomavirus model. Very few viral particles were detected in L2ATGko mutant-infected tissues. Interestingly, the localization of L1 in lesions induced by L2ATGko was primarily cytoplasmic rather than nuclear. The findings support the hypothesis that the L2 gene influences the expression, location, transport, and assembly of the L1 protein in vivo.


Subject(s)
Capsid Proteins/physiology , Mucous Membrane/virology , Oncogene Proteins, Viral/physiology , Papillomaviridae/physiology , Skin/virology , Animals , Capsid Proteins/genetics , Cell Transformation, Viral , DNA, Viral/biosynthesis , Female , Genome, Viral , Mice , Mice, Nude , Mutation , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , Virus Replication
16.
Tissue Barriers ; 9(4): 1943274, 2021 10 02.
Article in English | MEDLINE | ID: mdl-34241579

ABSTRACT

Oropharyngeal, airway, intestinal, and genital mucosal epithelia are the main portals of entry for the majority of human pathogenic viruses. To initiate systemic infection, viruses must first be transmitted across the mucosal epithelium and then spread across the body. However, mucosal epithelia have well-developed tight junctions, which have a strong barrier function that plays a critical role in preventing the spread and dissemination of viral pathogens. Viruses can overcome these barriers by disrupting the tight junctions of mucosal epithelia, which facilitate paracellular viral penetration and initiate systemic disease. Disruption of tight and adherens junctions may also release the sequestered viral receptors within the junctional areas, and liberation of hidden receptors may facilitate viral infection of mucosal epithelia. This review focuses on possible molecular mechanisms of virus-associated disruption of mucosal epithelial junctions and its role in transmucosal viral transmission and spread.


Subject(s)
Tight Junctions , Virus Diseases , Epithelium/virology , Humans , Mucous Membrane/virology , Tight Junctions/virology , Virus Diseases/transmission
17.
Vet Microbiol ; 260: 109182, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34315003

ABSTRACT

Immunosuppression can increase the susceptibility of chickens to other disease-causing pathogens and interfere with the efficacy of vaccination against those pathogens. Chicken anaemia virus (CAV) and infectious bursal disease virus (IBDV) are common causes of immunosuppression in chickens. Immunosuppression was induced by experimental infection with either CAV or IBDV to assess the effect of immunosuppression on the efficacy of vaccination with Mycoplasma gallisepticum strain ts-304 against infection with virulent M. gallisepticum, a common bacterial pathogen of chickens worldwide. Birds were experimentally infected with either CAV or IBDV at 1 week of age, before vaccination and challenge with M. gallisepticum to examine the effect of immunosuppression at the time of vaccination, or at 6 weeks of age, after vaccination against M. gallisepticum but before challenge with virulent M. gallisepticum, to investigate the effect of immunosuppression at the time of challenge. All birds were vaccinated with a single dose of the ts-304 vaccine at 3 weeks of age and experimentally challenged with the virulent M. gallisepticum strain Ap3AS at 8 weeks of age. In immunosuppressed chickens there was a reduction in protection offered by the ts-304 vaccine at two weeks after challenge, as measured by tracheal mucosal thicknesses, serum antibody levels against M. gallisepticum, air sac lesion scores and virulent M. gallisepticum load in the trachea. Immunosuppressed birds with detectable serum antibodies against M. gallisepticum were less likely to have tracheal lesions. This study has shown that immunosuppression caused by infection with CAV or IBDV can interfere with vaccination against mycoplasmosis in chickens.


Subject(s)
Birnaviridae Infections/veterinary , Chicken anemia virus/immunology , Chickens/immunology , Circoviridae Infections/veterinary , Infectious bursal disease virus/immunology , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/immunology , Poultry Diseases/prevention & control , Air Sacs/virology , Animals , Birnaviridae Infections/prevention & control , Birnaviridae Infections/virology , Chicken anemia virus/pathogenicity , Chickens/microbiology , Circoviridae Infections/prevention & control , Circoviridae Infections/virology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunosuppression Therapy/veterinary , Infectious bursal disease virus/pathogenicity , Mucous Membrane/virology , Mycoplasma Infections/microbiology , Mycoplasma Infections/prevention & control , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/microbiology , Trachea/virology
18.
PLoS Pathog ; 17(6): e1009632, 2021 06.
Article in English | MEDLINE | ID: mdl-34061907

ABSTRACT

Human immunodeficiency virus (HIV) vaccines have not been successful in clinical trials. Dimeric IgA (dIgA) in the form of secretory IgA is the most abundant antibody class in mucosal tissues, making dIgA a prime candidate for potential HIV vaccines. We coupled Positron Emission Tomography (PET) imaging and fluorescent microscopy of 64Cu-labeled, photoactivatable-GFP HIV (PA-GFP-BaL) and fluorescently labeled dIgA to determine how dIgA antibodies influence virus interaction with mucosal barriers and viral penetration in colorectal tissue. Our results show that HIV virions rapidly disseminate throughout the colon two hours after exposure. The presence of dIgA resulted in an increase in virions and penetration depth in the transverse colon. Moreover, virions were found in the mesenteric lymph nodes two hours after viral exposure, and the presence of dIgA led to an increase in virions in mesenteric lymph nodes. Taken together, these technologies enable in vivo and in situ visualization of antibody-virus interactions and detailed investigations of early events in HIV infection.


Subject(s)
Colon/virology , HIV Antibodies , HIV Infections , Immunoglobulin A, Secretory , Mucous Membrane/virology , Animals , Macaca mulatta , Mucous Membrane/immunology , Positron Emission Tomography Computed Tomography , Rectum
19.
Front Immunol ; 12: 625649, 2021.
Article in English | MEDLINE | ID: mdl-34093520

ABSTRACT

Genital mucosal transmission is the most common route of HIV spread. The initial responses triggered at the site of viral entry are reportedly affected by host factors, especially complement components present at the site, and this will have profound consequences on the outcome and pathogenesis of HIV infection. We studied the initial events associated with host-pathogen interactions by exposing cervical biopsies to free or complement-opsonized HIV. Opsonization resulted in higher rates of HIV acquisition/infection in mucosal tissues and emigrating dendritic cells. Transcriptomic and proteomic data showed a significantly more pathways and higher expression of genes and proteins associated with viral replication and pathways involved in different aspects of viral infection including interferon signaling, cytokine profile and dendritic cell maturation for the opsonized HIV. Moreover, the proteomics data indicate a general suppression by the HIV exposure. This clearly suggests that HIV opsonization alters the initial signaling pathways in the cervical mucosa in a manner that promotes viral establishment and infection. Our findings provide a foundation for further studies of the role these early HIV induced events play in HIV pathogenesis.


Subject(s)
Cervix Uteri/virology , Complement System Proteins/immunology , Gene Expression Profiling , HIV Infections/virology , HIV-1/pathogenicity , Mucous Membrane/virology , Proteome , Proteomics , Transcriptome , Cervix Uteri/immunology , Cervix Uteri/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Female , Gene Expression Regulation , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/growth & development , HIV-1/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate , Mucous Membrane/immunology , Mucous Membrane/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Time Factors , Tissue Culture Techniques , Virus Internalization , Virus Replication
20.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34156975

ABSTRACT

The ectocervix is part of the lower female reproductive tract (FRT), which is susceptible to sexually transmitted infections (STIs). Comprehensive knowledge of the phenotypes and T cell receptor (TCR) repertoire of tissue-resident memory T cells (TRMs) in the human FRT is lacking. We took single-cell RNA-Seq approaches to simultaneously define gene expression and TCR clonotypes of the human ectocervix. There were significantly more CD8+ than CD4+ T cells. Unsupervised clustering and trajectory analysis identified distinct populations of CD8+ T cells with IFNGhiGZMBloCD69hiCD103lo or IFNGloGZMBhiCD69medCD103hi phenotypes. Little overlap was seen between their TCR repertoires. Immunofluorescence staining showed that CD103+CD8+ TRMs were preferentially localized in the epithelium, whereas CD69+CD8+ TRMs were distributed evenly in the epithelium and stroma. Ex vivo assays indicated that up to 14% of cervical CD8+ TRM clonotypes were HSV-2 reactive in HSV-2-seropositive persons, reflecting physiologically relevant localization. Our studies identified subgroups of CD8+ TRMs in the human ectocervix that exhibited distinct expression of antiviral defense and tissue residency markers, anatomic locations, and TCR repertoires that target anatomically relevant viral antigens. Optimization of the location, number, and function of FRT TRMs is an important approach for improving host defenses to STIs.


Subject(s)
Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD8-Positive T-Lymphocytes/immunology , Cervix Uteri , Herpesvirus 2, Human , Integrin alpha Chains/analysis , Lectins, C-Type/analysis , Adaptive Immunity , CD4-Positive T-Lymphocytes/immunology , Cervix Uteri/immunology , Cervix Uteri/pathology , Cervix Uteri/virology , Female , Genes, T-Cell Receptor/immunology , Herpesvirus 2, Human/immunology , Herpesvirus 2, Human/isolation & purification , Humans , Immunologic Memory , Immunophenotyping/methods , Memory T Cells/immunology , Mucous Membrane/immunology , Mucous Membrane/pathology , Mucous Membrane/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...