Glennys V. Reynoso’s research while affiliated with National Institute of Allergy and Infectious Diseases, National Institutes of Health and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (51)


Figure 1. CHIKV RNA accumulates in MARCO-expressing floor LECs in the dLN. (A-F) WT mice were inoculated with PBS (mock, n = 2) or 10 3 PFU of CHIKV (n = 2) in the footpad. At 8 h post-infection, the dLN was collected and enzymatically digested into a single-cell suspension. Cells were enriched for CD45 -cells and analyzed by scRNA-seq. (A) UMAP projection shows CHIKV score, calculated as the fraction of total reads that align to the CHIKV genome for each cell from the CHIKV-enriched libraries. (B) UMAP projection shows CHIKV + cells. (C) CHIKV score is shown for CHIKV + cells for cell types with >40 total cells and >3 CHIKV + cells. P values were calculated using a one-sided Wilcoxon rank sum test with Bonferroni correction comparing each cell type with all other CHIKV + cells. Only adjusted p-values <0.05 are shown. (D) The fraction of cells identified as CHIKV + is shown for each cell type in C. P values were calculated using a one-sided hypergeometric test with Bonferroni correction. Labels show the number of
Figure 2. WT CHIKV infection disrupts LEC marker expression and elicits infiltration of LN sinuses. (A-D) WT mice were mock-inoculated (n = 3) or inoculated in the footpad with 10 3 PFU CHIKV 181/25 (n = 5) or WT CHIKV (n = 5) and the dLN was collected at 48 h post-infection. (A)
Figure 6. CHIKV-MARCO interactions promote LN inflammation. (A-E) WT and MARCO -/-mice were inoculated in the footpad with 10 3 PFU WT CHIKV (n = 9-13) or CHIKV E2 K200R (n = 10) (2-3 independent experiments). At the indicated timepoints, the dLN was collected for gene expression and viral RNA analysis by RT-qPCR (A-C) or for inflammatory myeloid cells by flow cytometry (D-F). (A) Expression of chemokines at 8, 12, and 16 h post-infection. (B) Expression
Figure 7. WT CHIKV infection impairs antigen acquisition by LECs in a MARCO-dependent manner. (A) WT and MARCO -/-mice were mock-inoculated (n = 5) or inoculated in the footpad with 10 3 PFU CHIKV 181/25 (n = 8) or WT CHIKV (n = 8) (2 independent experiments). At 72 h
Chikungunya virus infection disrupts lymph node lymphatic endothelial cell composition and function via MARCO
  • Article
  • Full-text available

January 2024

·

36 Reads

·

1 Citation

JCI Insight

Cormac J Lucas

·

Ryan M Sheridan

·

Glennys V Reynoso

·

[...]

·

Thomas E Morrison

Infection with chikungunya virus (CHIKV) causes disruption of draining lymph node (dLN) organization, including paracortical relocalization of B cells, loss of the B cell-T cell border, and lymphocyte depletion that is associated with infiltration of the LN with inflammatory myeloid cells. Here, we find that during the first 24 h of infection, CHIKV RNA accumulates in MARCO-expressing lymphatic endothelial cells (LECs) in both the floor and medullary LN sinuses. The accumulation of viral RNA in the LN was associated with a switch to an antiviral and inflammatory gene expression program across LN stromal cells, and this inflammatory response, including recruitment of myeloid cells to the LN, was accelerated by CHIKV-MARCO interactions. As CHIKV infection progressed, both floor and medullary LECs diminished in number, suggesting further functional impairment of the LN by infection. Consistent with this idea, we find that antigen acquisition by LECs, a key function of LN LECs during infection and immunization, was reduced during pathogenic CHIKV infection.

Download

Multifaceted roles for STAT3 in gammaherpesvirus latency revealed through in vivo B cell knockout models

January 2024

·

109 Reads

·

1 Citation

There are no directed therapies to the latency program of the human gammaherpesviruses, Epstein-Barr virus and Kaposi sarcoma herpesvirus. Activated host factor signal transducer and activator of transcription 3 (STAT3) is a hallmark of cancers caused by these viruses. We applied the murine gammaherpesvirus pathogen system to explore STAT3 function upon primary B cell infection in the host. Since STAT3 deletion in all CD19+ B cells of infected mice led to altered B and T cell responses, we generated chimeric mice with both normal and STAT3-deleted B cells. B cells lacking STAT3 failed to support virus latency compared to normal B cells from the same infected animal. Loss of STAT3 impaired B cell proliferation and differentiation and led to a striking upregulation of interferon-stimulated genes. These findings expand our understanding of STAT3-dependent processes that are key to its function as a pro-viral latency determinant for oncogenic gammaherpesviruses in B cells and may provide novel therapeutic targets.


Chikungunya virus infection disrupts lymph node lymphatic endothelial cell composition and function via MARCO

October 2023

·

3 Reads

Infection with chikungunya virus (CHIKV) causes disruption of draining lymph node (dLN) organization, including paracortical relocalization of B cells, loss of the B cell-T cell border, and lymphocyte depletion that is associated with infiltration of the LN with inflammatory myeloid cells. Here, we find that during the first 24 h of infection, CHIKV RNA accumulates in MARCO-expressing lymphatic endothelial cells (LECs) in both the floor and medullary LN sinuses. The accumulation of viral RNA in the LN was associated with a switch to an antiviral and inflammatory gene expression program across LN stromal cells, and this inflammatory response, including recruitment of myeloid cells to the LN, was accelerated by CHIKV-MARCO interactions. As CHIKV infection progressed, both floor and medullary LECs diminished in number, suggesting further functional impairment of the LN by infection. Consistent with this idea, we find that antigen acquisition by LECs, a key function of LN LECs during infection and immunization, was reduced during pathogenic CHIKV infection.


Type I Innate Lymphoid Cells Sense and Proliferate in Response to Distal Viral Infection

May 2023

·

10 Reads

The Journal of Immunology

Innate lymphoid cells (ILCs) are a diverse group of tissue-resident lymphocytes that lack antigen-specific receptors commonly found on adaptive immune cells. While many different types of ILCs have been described, only group 1 ILCs (ILC1s) are known to exhibit marked antiviral activity. We recently described ILC1-mediated protection of the oral mucosa, a critical barrier tissue and site of continued pathogen exposure. Based on these studies, we hypothesized that these innate antiviral cells might also respond to inflammation induced by a distal viral infection. To address this, we used a common mouse model of vaccinia virus (VACV) infection of the peritoneal cavity, which spreads locally but does not result in infection of the oral mucosa. Confocal microscopy of frozen sections of the lip demonstrated a marked increase in the number and density of oral mucosal ILC1s in response to distal infection. Further, ILC1s remained sustainably elevated for weeks following the initial infection. Ongoing experiments will assess the ability of this enhanced layer of ILC1s to provide heightened antiviral immunity in the oral mucosa and determine the timeframes in which this occurs. Together, our data suggest that the ability of ILC1s to globally sense infection and enhance protection in response to distal infection may represent a previously unappreciated mechanism of trained immunity in the tissue.


B cell-intrinsic STAT3-mediated support of latency and interferon suppression during murine gammaherpesvirus 68 infection revealed through an in vivo competition model

March 2023

·

24 Reads

·

2 Citations

Cancers associated with the oncogenic gammaherpesviruses, Epstein-Barr virus and Kaposi sarcoma herpesvirus, are notable for their constitutive activation of the transcription factor STAT3. To better understand the role of STAT3 during gammaherpesvirus latency and immune control, we utilized murine gammaherpesvirus 68 (MHV68) infection. Genetic deletion of STAT3 in B cells of CD19 cre/+ Stat3 f/f mice reduced peak latency approximately 7-fold. However, infected CD19 cre/+ Stat3 f/f mice exhibited disordered germinal centers and heightened virus-specific CD8 T cell responses compared to WT littermates. To circumvent the systemic immune alterations observed in the B cell-STAT3 knockout mice and more directly evaluate intrinsic roles for STAT3, we generated mixed bone marrow chimeras consisting of WT and STAT3-knockout B cells. Using a competitive model of infection, we discovered a dramatic reduction in latency in STAT3-knockout B cells compared to their WT B cell counterparts in the same lymphoid organ. RNA sequencing of sorted germinal center B cells revealed that STAT3 promotes proliferation and B cell processes of the germinal center but does not directly regulate viral gene expression. Last, this analysis uncovered a STAT3-dependent role for dampening type I IFN responses in newly infected B cells. Together, our data provide mechanistic insight into the role of STAT3 as a latency determinant in B cells for oncogenic gammaherpesviruses. IMPORTANCE There are no directed therapies to the latency program of the gammaherpesviruses, Epstein-Barr virus and Kaposi sarcoma herpesvirus. Activated host factor STAT3 is a hallmark of cancers caused by these viruses. We applied the murine gammaherpesvirus pathogen system to explore STAT3 function upon primary B cell infection in the host. Since STAT3 deletion in all CD19+ B cells of infected mice led to altered B and T cell responses, we generated chimeric mice with both normal and STAT3-deleted B cells. B cells lacking STAT3 failed to support virus latency compared to normal B cells from the same infected animal. Loss of STAT3 impaired B cell proliferation and differentiation and led to a striking upregulation of interferon-stimulated genes. These findings expand our understanding of STAT3-dependent processes key to its function as a pro-viral latency determinant for oncogenic gammaherpesviruses in B cells and may provide novel therapeutic targets.


Figure 2. ZIKV infects macrophages in LN sinuses (A) Confocal images of frozen PLN sections from nodes harvested 8 h p.i. from C57BL/6 mice (top panels), C57BL/6 mice given the anti-IFNAR1 Ab MAR1-5A3 (center panels), and Ifnar1 −/− mice (bottom panels). Blue, B cells; green, CD169 + macrophages; red, ZIKV E protein; white, CD31. The far right panels show a higher-magnification view of the SCS. Dashed ovals show specific areas with ZIKV E protein staining. An asterisk indicates patches of SSMs with E protein staining.
Figure 3. ZIKV disrupts LN macrophage networks (A) Flow plots generated from single-cell suspensions of PLNs harvested from uninfected C57BL/6 mice (left panel), ZIKV-infected C57BL/6 mice (center left), C57BL/6 + anti-IFNAR1 Ab MAR1-5A3 mice (center right), and Ifnar1 −/− mice (right) 72 h p.i. Cells were first gated on CD45 + B220 -CD3 -CD11b + CD11c low cells. Gating indicates SSMs (left gates, CD169 + F4/80 -) and MSMs (right gates, CD169 + F4/80 + ). (B) Frequency of CD169 + SSMs (far left and center right panels) or MSMs (center left and far right panels) in PLNs (blue bars) and ILNs (red bars) as a percentage of total LN
Figure 5. Migrating dendritic cells are dispensable for ZIKV dissemination after FP inoculation (A) Viral titers (FFUs per milliliter) in the PLN (left, blue bars), serum (center, gray bars), and FP (right, yellow bars) harvested 12 h p.i. from Siglec1-cre Ifnar fl/fl (homozygous Ifnar1 knockout in CD169 + cells [cKO]) mice, Siglec1-cre Ifnar fl/WT (heterozygous cKO) mice, and Ifnar1 −/− mice inoculated with 10 4 FFUs of ZIKV. The experiment was repeated 2 times with 3 mice per group. Results shown are pooled from two independent experiments. Dots represent individual mice (either pooled LNs or separate serum or feet) and the average of technical replicates. Dashed line, LOD for the assay. Values below the LOD are reported as half the LOD (125 FFU/mL). (B) Viral titers (FFUs per milliliter) in the PLN (left, blue bars), ILN (center, red bars), and serum (right, gray bars) harvested 12 h p.i. from C57BL/6 or Ccr7 −/− mice treated with the anti-IFNAR1 Ab MAR1-5A3 and inoculated with 10 4 FFUs of ZIKV. The experiment was
Figure 6. Monocytes are not needed for early ZIKV dissemination (A) Flow cytometry plots generated from single-cell suspensions of PLNs harvested from naive C57BL/6 mice (left) and infected C57BL/6 mice at 24 h p.i. (all other panels). Where indicated, mice were given the MAR1-5A3 (anti-IFNAR1) or GR1 Ab before infection. An additional dose of MAR1-5A3 was given at the time of infection. Cells were first gated on CD45 + cells. Gating indicates Ly6C + monocytes (top boxes) and Ly6G + neutrophils (center boxes).
Figure 7. CD169 + macrophage infection does not result in morbidity (A) Viral titers (FFUs per milliliter) in the PLN (left, blue dots) and serum (right, gray bars) harvested at the indicated day p.i. from Ifnar1 −/− mice. The experiment was repeated 2 times with 3-4 mice per group. Results shown are pooled from two experiments. Dots represent individual mice (either pooled LNs or separate serum) and the average of technical replicates. Dashed line, LOD for the assay. Values below the LOD are reported as half the LOD (125 FFU/mL). (B) As in (A) but in Siglec1-cre Ifnar fl/fl (cKO) mice.
Zika virus spreads through infection of lymph node-resident macrophages

February 2023

·

62 Reads

·

18 Citations

Cell Reports

To disseminate through the body, Zika virus (ZIKV) is thought to exploit the mobility of myeloid cells, in particular monocytes and dendritic cells. However, the timing and mechanisms underlying shuttling of the virus by immune cells remains unclear. To understand the early steps in ZIKV transit from the skin, at different time points, we spatially mapped ZIKV infection in lymph nodes (LNs), an intermediary site en route to the blood. Contrary to prevailing hypotheses, migratory immune cells are not required for the virus to reach the LNs or blood. Instead, ZIKV rapidly infects a subset of sessile CD169+ macrophages in the LNs, which release the virus to infect downstream LNs. Infection of CD169+ macrophages alone is sufficient to initiate viremia. Overall, our experiments indicate that macrophages that reside in the LNs contribute to initial ZIKV spread. These studies enhance our understanding of ZIKV dissemination and identify another anatomical site for potential antiviral intervention.


The ectromelia virus virulence factor C15 facilitates early viral spread by inhibiting NK cell contact

November 2022

·

16 Reads

·

3 Citations

iScience

The success of poxviruses as pathogens depends upon their antagonism of host responses by multiple immunomodulatory proteins. The largest of these expressed by ectromelia virus (the agent of mousepox) is C15, one member of a well-conserved poxviral family previously shown to inhibit T cell activation. Here, we demonstrate by quantitative immunofluorescence imaging that C15 also limits contact between natural killer (NK) cells and infected cells in vivo. This corresponds to an inhibition in the number of total and degranulating NK cells, ex vivo and in vitro, with no detectable impact on NK cell cytokine production nor the transcription of factors related to NK cell recruitment or activation. Thus, in addition to its previously identified capacity to antagonize CD4 T cell activation, C15 inhibits NK cell cytolytic function, which results in increased viral replication and dissemination in vivo. This work builds on a body of literature demonstrating the importance of early restriction of virus within the draining lymph node.


Figure 1. Fungal recognition variants in patients with DCM. (A) Fold-enrichment of CLEC7A, c.714T>G; p.Y238* variant in patients with DCM compared with the gnomAD. Normalized to frequency of homozygous WT or variant carriers in gnomAD. P = 0.0303, Fisher's exact test. (B) Parallel signaling pathways after β-glucan recognition by DECTIN-1 leading to activation of NF-κB and NFAT transcription factors and production of TNF-α. The figure was created using BioRender. (C) Confocal microscopy of lung from C. posadasii-infected C57BL/6 mouse showing DECTIN-1 (red) localized near endospores (blue) (left), LAMP-1 (green) localization near endospores (middle), and colocalization of DECTIN-1 and LAMP-1 (tan) around endospores (right). (D) Frequency of patients of European ancestry with DCM with PLCG2, c.802C>T; p.R268W genotype normalized to the non-Finnish European population in gnomAD. P = 0.0077 Fisher's exact test. (E) Particulate β-glucaninduced TNF production by PBMCs from patients (n = 16) or healthy control (HC) participants (n = 12). DECTIN-1 variants (n = 4) include homozygous p.Y238* (filled) and heterozygous p.Y238* (open). Patients withPLCG2 variants (n = 6) include p.R268W heterozygotes (yellow symbols), p.M28L heterozygotes (green symbol), and p.R268W and p.K775R compound heterozygotes (open yellow symbol). Patients in the "other" category (n = 6) lack identified causal variants. P values were calculated using Brown-Forsythe and Welch ANOVA with Dunnett's T3 multiple comparisons test. (F) Frequency of Y238* among East Asian patients from DCM validation cohort compared with the 1000G and gnomAD.
Figure 2. DECTIN-1 signaling drives H 2 O 2 production by DUOX1 and DUOXA1 in HEK cells. (A) DECTIN-1-activated PLCγ2 releases intracellular Ca ++ , which activates the EF-hand domains of DUOX1, leading to H 2 O 2 production. DUOX1 and DUOXA1 patient variants are highlighted by yellow stars. (B) DUOX1 variants identified in patients with DCM were transfected into HEK Flp-In cells stably expressing WT DUOXA1. Cells were stimulated with ionomycin, and H 2 O 2 production was measured for 60 minutes. Results are the average of triplicate wells presented as the ratio of H 2 O 2 production by variant or WT; each dot represents the average of triplicate wells from a unique experiment. ****P < 0.0001 by ordinary 1-way ANOVA and Dunnett's multiple comparisons test. Western blot showing decreased protein abundance of several DUOX1 variants after transfection. (C) DUOXA1 variants, identified in patients with DCM, transfected into HEK Flp-In cells stably expressing DUOX1. Measured as in B. P = 0.016 by ordinary 1-way ANOVA and Dunnett's multiple comparisons test. (D) Hydrogen peroxide production in HEK cells transfected with WT or patient variant DUOX1, DUOXA1, or DECTIN1, or lacking PLCG2 constructs. Results are the average of triplicate wells presented as the ratio of H 2 O 2 production by variant/WT; each dot represents a unique experiment. ****P < 0.0001 using ordinary 1-way ANOVA and Dunnett's multiple comparisons test.
Figure 3. Exploratory cohort: 34 patients with DCM with identified fungal pattern recognition pathway or DUOX1 / DUOXA1 variants.
DCM patients with additional infections and identified variants
Immunogenetics associated with severe coccidioidomycosis

September 2022

·

161 Reads

·

29 Citations

JCI Insight

Disseminated coccidioidomycosis (DCM) is caused by Coccidioides, pathogenic fungi endemic to the Southwestern United States and Mexico. Illness occurs in approximately 30% of those infected, <1% of whom develop disseminated disease. To address why some individuals allow dissemination, we enrolled DCM patients and performed whole-exome sequencing. In an exploratory set of 67 DCM patients, two had haploinsufficient STAT3 mutations, while defects in β-glucan sensing and response were seen in 34/67 (50.7%) cases. Damaging CLEC7A (n=14) and PLCG2 (n=11) variants were associated with impaired production of β-glucan-stimulated TNF-α from peripheral blood mononuclear cells compared to healthy controls (P<0.005). Using ancestry-matched controls, damaging CLEC7A and PLCG2 variants were over-represented in DCM (P=0.0206, P=0.015, respectively) including CLEC7A Y238* (P=0.0105) and PLCG2 R268W (P=0.0025). A validation cohort of 111 DCM patients confirmed PLCG2 R268W (P=0.0276), CLEC7A I223S (P=0.044), and CLEC7A Y238* (P=0.0656). Stimulation with a DECTIN-1 agonist induced DUOX1/DUOXA1-derived H2O2 in transfected cells. Heterozygous DUOX1 or DUOXA1 variants which impaired H2O2 production were overrepresented in discovery and validation cohorts. Patients with DCM have impaired β-glucan sensing or response affecting TNF-α and H2O2 production. Impaired Coccidioides recognition and decreased cellular response are associated with disseminated coccidioidomycosis.


Figure 1: ZIKV replicates in the PLN before systemic dissemination (A) Viral titers (in focus-forming units (FFU)/ml)) in the PLN (left, blue dots) or serum (right, grey dots) harvested at the indicated time (in min) during the first hr following footpad (FP) injection of Ifnar1 -/-mice with 10 4 FFU of ZIKV H/PF/2013. Dots represent individual mice (either pooled PLNs or separate sera) and the average of technical replicates in the focus-forming assay (FFA). A dashed line shows the limit of detection of the FFA. Values below half the LOD are
Figure 4: Lymph node macrophage infection alone supports systemic dissemination
Figure 5: Migrating dendritic cells are dispensable for ZIKV dissemination. (A) Viral titers (FFU/ml) in the PLN (left, blue bars), serum (middle, grey bars), or footpad (right, yellow bars) harvested 12 hr p.i. from Siglec1-cre Ifnar fl/fl (homozygous Ifnar1 knockout in CD169 + cells, cKO) mice, Siglec1-cre Ifnar fl/WT (heterozygous cKO mice) or Ifnar1 -/-mice with 10 4 FFU of ZIKV. Experiment was repeated 2 times with 3 mice per group. Results shown are pooled from two independent experiments. Dots represent individual mice (either pooled LNs or separate serum or feet) and the average of technical replicates (values of zero are not shown due to log scale). Dashed line = limit of detection (LOD) for assay. Values below half the LOD are reported as half the LOD (125 FFU/ml). (B) Viral titers (FFU/ml) in the PLN (left, blue bars), ILN (middle, red bars), or serum (right, grey bars) harvested 12 hr p.i. from C57BL/6 or Ccr7 -/-mice treated with MAR1 and infected with 10 4 FFU of ZIKV. Experiment was repeated 2 times with 3 mice per group. Results shown are pooled from two independent experiments. Dots represent individual mice (either pooled LNs or separate serum) and the average of technical replicates (values of zero are not shown due to log scale). Dashed line = limit of detection for assay. Values below half the LOD are reported as half the LOD (125 FFU/ml). (C) Frequency of CD169 + SSMs (left) or MSMs (right) in the either PLNs (blue bars) or ILNs (red bars) 72 hr p.i. of C57BL/6 or Ccr7 -/-mice as a percentage of total LN macrophages Statistics = One-way
Figure 6: Monocytes are not needed for early ZIKV dissemination (A) Flow plots generated from single cell suspensions of PLNs harvested from naïve C57BL/6 mice (left) or infected C57BL/6 mice at 24 hr p.i. (all other panels). Where indicated, mice were given MAR1 or Gr1 Ab before infection. An additional dose of MAR1 was given at the time of infection. Cells were first gated on CD45 + cells. Gating indicates Ly6c + monocytes (top boxes) and Ly6g + neutrophils (middle boxes). (B) Frequency of Ly6c + monocytes in the blood (left) or PLN (right) of indicated mice at 24 hr p.i. Statistics = One-way ANOVA. Dots show pooled LNs from individual mice. Scalebars = SEM. Experiment was repeated 2 times with 3-5 mice/group. (C) Confocal images of frozen PLN sections from nodes harvested 24 hr p.i. from LysM-eGFP mice treated with MAR1. B220 = blue; LysM-GFP + cells = green; blood vessel (CD31) = white; and ZIKV NS2b protein = red. Right panels omit B cells for clarity. Scalebars = µms. (D) Viral
Figure 7: Nodal macrophage infection does not result in morbidity
Zika virus spreads through infection of lymph node-resident macrophages

September 2022

·

83 Reads

·

1 Citation

Zika virus (ZIKV) is an arthopod-vectored flavivirus that disseminates from the infection site into peripheral tissues, where it can elicit virus-induced pathology. To move through the body, ZIKV is thought to exploit the mobility of myeloid cells, in particular monocytes and dendritic cells. However, multiple distinct steps during viral spread culminate in peripheral tissue infection, and the timing and mechanisms underlying mobile immune cell shuttling of virus remain unclear. To understand the very early steps in ZIKV dissemination from the skin, we kinetically and spatially mapped ZIKV-infected lymph nodes (LNs), an intermediary stop en route to the blood. Contrary to dogma, migratory immune cells were not required for large quantities of virus to reach the LN or blood. Instead, ZIKV rapidly infected a subset of immobile macrophages in the LN, which shed virus through the lymphatic pathway into the blood. Importantly, infection of LN macrophages alone was sufficient to initiate viremia. Together, our studies indicate that sessile macrophages that live and die in the LN contribute to initial ZIKV spread to the blood. These data build a more complete picture of ZIKV movement through the body and identify an alternate anatomical site for potential antiviral intervention. Highlights ZIKV infects and replicates in distinct LN macrophage populations LN macrophage infection results in infectious virus in the blood Virus reaches the blood in the absence of DC migration or monocyte infection Nodal macrophage infection does not sustain viremia or induce morbidity Graphical abstract Caption ZIKV infects lymph node macrophages, which shed infectious virus into the lymph and then blood.


The ectromelia virus virulence factor C15 facilitates early viral spread by inhibiting NK cell-infected cell contacts

June 2022

·

6 Reads

The success of poxviruses as pathogens depends upon their antagonism of host responses by multiple immunomodulatory proteins. The largest of these expressed by ectromelia virus (the agent of mousepox) is C15, one member of a well-conserved poxviral family previously shown to inhibit T cell activation. Here, we demonstrate by quantitative immunofluorescence imaging that C15 also limits contact between natural killer (NK) cells and infected cells in vivo . This corresponds to an inhibition in the number of total and degranulating NK cells, ex vivo and in vitro , with no detectable impact on NK cell cytokine production nor the transcription of factors related to NK cell recruitment or activation. Thus, in addition to its previously identified capacity to antagonize CD4 T cell activation, C15 inhibits NK cell cytolytic function, which results in increased viral replication and dissemination in vivo . This work builds on a body of literature demonstrating the importance of early restriction of virus within the draining lymph node. Summary Poxvirus B22 family proteins are important virulence factors known to inhibit T cell functions. Peauroi et al. identify a novel function of the ectromelia virus homolog, C15, which inhibits NK cell-target contact and cytolytic function to facilitate early viral spread. (Provide a short, ∼40-word summary statement for the online JEM table of contents and alerts. This summary should describe the context and significance of the findings for a general readership; it should be written in the present tense and refer to the work in the third person.)


Citations (24)


... In addition to acquisition and retention of foreign protein antigens, LECs may support viral replication, such as Kaposi's sarcoma associated herpes virus 15 . Recently, single cell mRNA sequencing of LNSCs during CHIKV infection indicated that subsets of LECs that express the scavenger receptor MARCO may support CHIKV RNA replication 16 , consistent with another study showing MARCO-dependent internalization of CHIKV by LN LECs 17 . CHIKV RNA is also detectable in FRCs, which express the CHIKV entry receptor Mxra8 18,19 . ...

Reference:

A specific and portable gene expression program underlies antigen archiving by lymphatic endothelial cells
Chikungunya virus infection disrupts lymph node lymphatic endothelial cell composition and function via MARCO

JCI Insight

... STAT3 is important for the establishment of longterm latency by MHV68 [92]. However, in contrast to our findings for EBV, STAT3 does not directly regulate MHV68 viral gene expression, but instead dampens type I IFN responses in newly infected B-cells [93]. Thus, EBV has evolved specific mechanisms to coopt B-cell STAT signaling to modulate latency gene expression in response to B-cell cues. ...

Multifaceted roles for STAT3 in gammaherpesvirus latency revealed through in vivo B cell knockout models

... Neutralization was tested by means of a plaque reduction neutralization test (PRNT) as described 84 . Briefly, three-fold serum dilutions, starting from an initial concentration of 1:80 in cDMEM were incubated with 150 PFU of MHV68 on ice for one hour. ...

B cell-intrinsic STAT3-mediated support of latency and interferon suppression during murine gammaherpesvirus 68 infection revealed through an in vivo competition model

... As LPS, IFN-γ, and LPS + IFN-γ exposure induced clearly differentiable pro-inflammatory subsets through random forest modeling, we sought to classify gene expression patterns of macrophages exposed to various bacterial and viral agents as M(LPS)-, M(IFN-γ)-, or M(LPS + IFN-γ)-like. Most importantly, while HIV 108 and the Zika virus 109,110 are known to infect both monocytes and macrophages, we found that virally exposed macrophages resembled unpolarized M0 or M(LPS + IFN-γ) macrophages, but not M(LPS) or M(IFN-γ) macrophages (Fig. 6). Additionally, Influenza A virus (IAV), which demonstrates strain and macrophage lineagespecific replicative properties 111 , induce states predicted as M0 cells (Fig. 6), suggesting no polarization-like response is elicited following exposure. ...

Zika virus spreads through infection of lymph node-resident macrophages

Cell Reports

... Although the top BLASTP hits for ORF65 are for related proteins in ECIV and SDDV, the majority of BLASTP hits are to B22 proteins in poxviruses. B22 proteins are typically found in chordopoxvirus genomes and have been determined to inhibit antigen presentation to T-lymphocytes and natural killer cells during infections, increasing poxvirus virulence [50][51][52]. The two outer circles represent predicted ORFs colored based on shared homology with proteins from other iridoviruses, poxviruses, or eukaryotic organisms. ...

The ectromelia virus virulence factor C15 facilitates early viral spread by inhibiting NK cell contact

iScience

... Otherwise, he did not have the connective tissue abnormalities typically seen in patients with STAT3 DN variants [59]. Similarly, two other patients had unremarkable histories until the development of disseminated coccidioidomycosis, which was eventually fatal due to central nerve system involvement [60]. ...

Immunogenetics associated with severe coccidioidomycosis

JCI Insight

... Whether this capture was receptor-mediated or promiscuous was not determined. In addition, LECs expressing the scavenger receptor MARCO but not CD169 + macrophages were shown to contribute to controlling Chikungunya virus and other alphaviruses in the dLN [43]. Notably, Reynoso et al. showed that wild-type VACV inoculated into the footpad at high doses (10 6 -10 8 pfu) passed through the SCS and entered dLN conduits as free virions to infect paracortical DCs one hour post-infection [44]. ...

MARCO+ lymphatic endothelial cells sequester arthritogenic alphaviruses to limit viremia and viral dissemination
  • Citing Article
  • October 2021

The EMBO Journal

... Helminth infection elicits type 2 immune responses, which are largely dependent on the transcription factor STAT6 (39). In coinfection studies, STAT6 signaling is a primary pathway by which enteric helminth infection hinders antiviral T cell immunity (40,41). To investigate whether STAT6 signaling mediates Hpb-triggered defects in vaccine-generated CD8 + T cell responses, we repeated Hpb infection and mRNA vaccination in Stat6 −/− and congenic wild-type (WT) mice. ...

Enteric helminth coinfection enhances host susceptibility to neurotropic flaviviruses via a tuft cell-IL-4 receptor signaling axis
  • Citing Article
  • February 2021

Cell

... The development of LCs in the vicinity of the dental plaque is enhanced by the microbiota 11,14 , supporting the stronger interaction of the buccal gingiva with the microbiota. The elevated levels of CD8 + T cells and ILCs in the buccal gingival epithelium of SPF mice also align with this notion, as both cell types have been shown to play a role in preventing viral infection in the oral mucosa 49,50 . This implies that the buccal and palatal gingival epithelia constitute immunologically distinct regions, potentially influencing pathological processes within this oral barrier. ...

Group 1 innate lymphoid-cell-derived interferon-γ maintains anti-viral vigilance in the mucosal epithelium

Immunity

... We previously showed responses to Modi-2 peptides in the CpG/ MPLA adjuvant formulation were CD4-mediated. However, the SNAPvax formulation has been shown to enhance the potency of peptide-based vaccines that mainly induced CD8 T cells responses 21,24 . Therefore, we reinvestigated whether responses to Modi-2 SNAPvax formulation were CD4 or CD8 mediated. ...

Intravenous nanoparticle vaccination generates stem-like TCF1 neoantigen-specific CD8 T cells

Nature Immunology