[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (986)

Search Parameters:
Keywords = cytomegalovirus

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 693 KiB  
Communication
Gene Therapy in the Light of Lifestyle Diseases: Budesonide, Acetaminophen and Simvastatin Modulates rAAV Transduction Efficiency
by Żaneta Słyk, Natalia Stachowiak and Maciej Małecki
Pharmaceuticals 2024, 17(9), 1213; https://doi.org/10.3390/ph17091213 - 14 Sep 2024
Viewed by 283
Abstract
Recombinant AAV (rAAV) vectors are increasingly favored for gene therapy due to their useful features of vectorology, such as transfection of dividing and nondividing cells, the presence of tissue-specific serotypes, and biosafety considerations. This study investigates the impact of commonly used therapeutic drugs—acetaminophen, [...] Read more.
Recombinant AAV (rAAV) vectors are increasingly favored for gene therapy due to their useful features of vectorology, such as transfection of dividing and nondividing cells, the presence of tissue-specific serotypes, and biosafety considerations. This study investigates the impact of commonly used therapeutic drugs—acetaminophen, budesonide, and simvastatin—on rAAV transduction efficiency in HEK-293 cells. Cells were transduced with an AAV mosaic vector under the control of a cytomegalovirus (CMV) promoter encoding green fluorescent protein (GFP). Transduction efficiency was assessed by qPCR and fluorescent microscopy. Analysis of functional interactions between genes potentially involved in rAAV transduction in drug-exposed cells was also performed. This study showed a clear effect of drugs on rAAV transmission. Notably, acetaminophen enhanced transduction efficiency by 9-fold, while budesonide and simvastatin showed 2-fold and 3-fold increases, respectively. The gene analysis illustrates the possible involvement of genes related to cell membranes in the potentiation of rAAV transduction induced by the drugs under investigation. Attention should be paid to S100A8, which is a common drug-modified gene for drugs showing anti-inflammatory effects (budesonide and simvastatin), demonstrating an interaction with the gene encoding the receptor for AAV (HGFR). This study underscores the significance of assessing rAAV pharmacokinetics/pharmacodynamics (PKs/PDs) and drug–gene therapy interactions in optimizing gene therapy protocols. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

Figure 1
<p>AAV/DJ transduction efficiency of the HEK-293 cell line in the presence of budesonide (gray bars), acetaminophen (dark blue bars), and simvastatin (light blue bars). Results are presented as the genome copy number ± standard deviation. The control group (red bar) was the HEK-293 cells transduced with rAAV/DJ alone. The area plot shows the fold change in transduction efficiency of HEK-293 cells with the rAAV/DJ vector in the presence of drugs compared with the control and is expressed as a percentage.</p>
Full article ">Figure 2
<p>Transduction of HEK-293 cell line with rAAV/DJ at an extremely low MOI (300) in the presence of budesonide (gray color), acetaminophen (dark blue color), and simvastatin (light blue color). BF—bright field, FITC—fluorescent field, scale bar—50 µm. The control group (red color) includes samples transduced with rAAV/DJ without drugs. The column graph shows the number of GFP-positive (GFP+) cells as a fraction of the total number of cells present in the field of view. The area plot shows the fold change in the transduction efficiency of HEK-293 cells with the rAAV/DJ vector in the presence of drugs compared to the control and is expressed as a percentage.</p>
Full article ">Figure 3
<p>Correlation analysis between rAAV transmission genes (red nodes) and drug-modified genes (gray, dark blue, or light blue for budesonide, acetaminophen, or simvastatin, respectively) using a STRING database [<a href="#B4-pharmaceuticals-17-01213" class="html-bibr">4</a>]. The obtained hits were divided into selected cellular compartments (plasma membrane, cytosol, and nucleus) presented horizontally in the graph. The hit score constitutes the combined score &gt; 0.9, which is computed by combining the probabilities from the different evidence channels and correcting for the probability of randomly observing an interaction.</p>
Full article ">Figure 4
<p>Expression of rAAV receptors, KIA319LL and SDC2, in HEK-293 cells after treatment with budesonide, acetaminophen, and simvastatin. The statistical significance was analyzed between the samples treated with medicines (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.0001, and **** <span class="html-italic">p</span> &lt; 0.00001) using a Kruskal–Wallis test and a subsequently performed post hoc test, Dunn’s test.</p>
Full article ">
11 pages, 1587 KiB  
Article
Different Antigen-Specific CD4+ and CD8+ T-Cell Response against HCMV Proteins in Pregnant Women with Primary Infection and in Control Subjects with Remote Infection
by Federica Zavaglio, Piera d’Angelo, Chiara Fornara, Paola Zelini, Giuditta Comolli, Milena Furione, Alessia Arossa, Arsenio Spinillo, Daniele Lilleri and Fausto Baldanti
J. Clin. Med. 2024, 13(18), 5448; https://doi.org/10.3390/jcm13185448 - 13 Sep 2024
Viewed by 272
Abstract
Background/Objectives: Human cytomegalovirus (HCMV) is the most frequent cause of congenital infections. The HCMV-specific T-cell response in primary infection may help define reliable correlates of immune protection in pregnancy. In this study, the antigen-specific T-cell response against different HCMV proteins (IE-1, pp65, [...] Read more.
Background/Objectives: Human cytomegalovirus (HCMV) is the most frequent cause of congenital infections. The HCMV-specific T-cell response in primary infection may help define reliable correlates of immune protection in pregnancy. In this study, the antigen-specific T-cell response against different HCMV proteins (IE-1, pp65, gB, gHgLpUL128L) was investigated in pregnant women with primary infection and in control subjects with remote infection to identify possible components of a vaccine. Methods: Blood samples from 35 pregnant women with HCMV primary infection and 30 HCMV-seropositive healthy adult subjects with remote infection were tested. The antigen-specific T-cell response was measured using cytokine intracellular staining after stimulation with IE-1, pp65, gB and gHgLpUL128L peptides pool. Results: The pp65-specific CD4+ T-cell response was higher in pregnant women with HCMV primary infection at the late time point and in control subjects with remote infection, while the pregnant women at the early time point showed a higher gB-specific CD8+ T-cell response. Regarding the CD4+ and CD8+ T-cell phenotypes, we observed that HCMV-specific CD4+ and CD8+ T cells expressing CD45RA+ remained constant in pregnant women with primary infection at the early and late time points and in subjects with remote infection, while HCMV-specific CD4+ and CD8+ T cells expressing IL-7R+ or producing IL-2 were higher in control subjects with remote infection than in pregnant women with HCMV primary infection. Conclusions: The T-cell response was higher against gB in the early phase of infection and against pp65 in the late phase. Therefore, these proteins should be taken into consideration as candidates for a vaccine. Full article
(This article belongs to the Section Immunology)
Show Figures

Figure 1

Figure 1
<p>Percentage of HCMV-specific T cells producing IFN-γ after stimulation with IE-1, pp65, gB, gHgLpUL128L in pregnant women with HCMV primary infection at the early and the late time points, and in control subjects with HCMV remote infection. (<b>A</b>) Antigen-specific IFN-γ<sup>+</sup> CD4<sup>+</sup> T cells and (<b>B</b>) IFN-γ<sup>+</sup> CD8<sup>+</sup> T cells. Early time point: median: 60; (IQR49-65) days after onset infection. Late time point: median: 360; (IQR 356-412) days after onset infection. *** <span class="html-italic">p</span> &lt; 0.001. Dash lines divide the different peptides pool.</p>
Full article ">Figure 2
<p>Frequency of CD4<sup>+</sup> and CD8<sup>+</sup> T-cell responders to HCMV peptide pools (IE-1, pp65, gB and gHgLpUL128L) detected in pregnant women with HCMV primary at the early and the late time point and in control subjects with HCMV remote infection. Percentage of responders to different numbers HCMV peptides pool (<b>A</b>,<b>B</b>) and frequencies of responders to IE-1, pp65, gB and gHgLpUL128L peptides pool (<b>C</b>,<b>D</b>), are reported. Early time point: median: 60; (IQR 49–65) days after onset infection. Late time point: median: 360; (IQR 356–412) days after onset infection. Dash lines divide the different peptides pool.</p>
Full article ">Figure 3
<p>IE1, pp65, gB and gHgLpUL128L-specific CD4<sup>+</sup> and CD8<sup>+</sup> T cells expressing (<b>A</b>,<b>B</b>) CD45RA<sup>+</sup>, (<b>C</b>,<b>D</b>) IL-7R<sup>+</sup> and (<b>E</b>,<b>F</b>) producing IL-2 in pregnant women with HCMV primary infection at the early and late time point and in subjects with remote infection. Early time point: median: 60; (IQR 49–65) days after onset infection. Late time point: median: 360; (IQR 356–412) days after onset infection. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. Dash lines divide the different peptides pool.</p>
Full article ">Figure 4
<p>CD4<sup>+</sup> and CD8<sup>+</sup> T-cell response to human cytomegalovirus (HCMV) peptides pool proteins (IE-1, pp65, gB and gHgLpUL128L) detected in 24 non-transmitting and 11 transmitting pregnant women at the early time point. (<b>A</b>) IFNγ<sup>+</sup> CD4<sup>+</sup> T cells. (<b>B</b>) IFNγ<sup>+</sup> CD8<sup>+</sup> T cells. Early time point: median: 60; (IQR 49–65) days after onset infection.</p>
Full article ">
9 pages, 434 KiB  
Article
Long-Term Follow Up of Patients Treated for Inflammatory Bowel Disease and Cytomegalovirus Colitis
by Gurtej Singh, Clarissa Rentsch, William Beattie, Britt Christensen, Finlay Macrae and Jonathan P. Segal
Diagnostics 2024, 14(18), 2030; https://doi.org/10.3390/diagnostics14182030 - 13 Sep 2024
Viewed by 301
Abstract
Background: Pathological reactivation of latent Cytomegalovirus (CMV) is triggered by inflammation and immunosuppression; both present in the pathogenesis and treatment of Inflammatory Bowel Disease (IBD). Whether CMV reactivation is associated with escalating medical therapy, further hospital admissions, or worse clinical outcomes remains controversial. [...] Read more.
Background: Pathological reactivation of latent Cytomegalovirus (CMV) is triggered by inflammation and immunosuppression; both present in the pathogenesis and treatment of Inflammatory Bowel Disease (IBD). Whether CMV reactivation is associated with escalating medical therapy, further hospital admissions, or worse clinical outcomes remains controversial. This study aimed to follow up IBD patients with an index episode of CMV colitis and analyse the clinical outcomes. Methods: A retrospective study of patients with IBD treated for CMV colitis was completed. The outcome results were collected at 6-month and 12-month time points after the first episode of CMV colitis. A total of 13 patients with Ulcerative Colitis and 1 with Crohn’s Disease were included. Results: CMV colitis recurrence occurred in 29% of patients at 12 months. A total of 43% of patients had changed their biologic dose at 6 months and 29% had escalated their biologic dose at 12 months. At 12 months, 36% of patients had been re-hospitalised, including three colectomies. Disease remission was only achieved by 29% of patients at 12 months. Conclusions: IBD patients with CMV colitis have substantial rates of re-hospitalisation, failed medical therapy, and colectomy. These risks may be greater at <6 months from an index episode of CMV colitis. Full article
(This article belongs to the Special Issue Inflammatory Pathologies)
Show Figures

Figure 1

Figure 1
<p>Consort flowchart showing inclusion and exclusion of patients included in final analysis.</p>
Full article ">
24 pages, 17687 KiB  
Article
Secondary Envelopment of Human Cytomegalovirus Is a Fast Process Utilizing the Endocytic Compartment as a Major Membrane Source
by Tim Bergner, Laura Cortez Rayas, Gesa Freimann, Clarissa Read and Jens von Einem
Biomolecules 2024, 14(9), 1149; https://doi.org/10.3390/biom14091149 - 12 Sep 2024
Viewed by 313
Abstract
Secondary envelopment of the human cytomegalovirus (HCMV) is a critical but not well-understood process that takes place at the cytoplasmic viral assembly complex (cVAC) where nucleocapsids acquire their envelope by budding into cellular membranes containing viral glycoproteins. Previous studies presented controversial results regarding [...] Read more.
Secondary envelopment of the human cytomegalovirus (HCMV) is a critical but not well-understood process that takes place at the cytoplasmic viral assembly complex (cVAC) where nucleocapsids acquire their envelope by budding into cellular membranes containing viral glycoproteins. Previous studies presented controversial results regarding the composition of the viral envelope, suggesting trans-Golgi and endosomal origins, as well as intersections with the exosomal and endocytic pathways. Here, we investigated the role of endocytic membranes for the secondary envelopment of HCMV by using wheat germ agglutinin (WGA) pulse labeling to label glycoproteins at the plasma membrane and to follow their trafficking during HCMV infection by light microscopy and transmission electron microscopy (TEM). WGA labeled different membrane compartments within the cVAC, including early endosomes, multivesicular bodies, trans-Golgi, and recycling endosomes. Furthermore, TEM analysis showed that almost 90% of capsids undergoing secondary envelopment and 50% of enveloped capsids were WGA-positive within 90 min. Our data reveal extensive remodeling of the endocytic compartment in the late stage of HCMV infection, where the endocytic compartment provides an optimized environment for virion morphogenesis and serves as the primary membrane source for secondary envelopment. Furthermore, we show that secondary envelopment is a rapid process in which endocytosed membranes are transported from the plasma membrane to the cVAC within minutes to be utilized by capsids for envelopment. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Viral Infections)
Show Figures

Figure 1

Figure 1
<p>Wheat germ agglutinin (WGA) labels the endocytic compartment. Uninfected fibroblasts were labeled with WGA coupled to horse radish peroxidase (WGA-HRP) for a 60-min pulse and a 30-min chase, both at 37 °C. (<b>A</b>) Overview of a cell with corresponding details shown in panels 1–4. (<b>1</b>,<b>2</b>) DAB precipitate appears dark and labels WGA-positive membranes distributed throughout the cytoplasm. (<b>3</b>,<b>4</b>) Detailed depiction of DAB precipitate. Note its localization at the luminal face of intercellular membrane compartments (arrowheads). (<b>3</b>) Large WGA-positive endosomes are located close to the cell surface, often containing small intraluminal vesicles (=multivesicular bodies (MVBs), white asterisks). (<b>4</b>) Vesicles of the endocytic compartment with different sizes (arrows) can be found close to trans-Golgi membranes. WGA-positive membranes were also part of Golgi cisternae (black asterisks), either at the terminal regions of the trans-most Golgi cisternae or (<b>B</b>) decorating the entire cross-section of the trans-most cisternae. Cy cytoplasm, Nu nucleus. Scale bars, 500 nm.</p>
Full article ">Figure 2
<p>Uptake of WGA-FITC into early endosomes in uninfected cells. Immunofluorescence microscopy of fibroblasts labeled with WGA-FITC (10 µg/mL) with a 60-min pulse and a 30-min chase, both at 37 °C. (<b>A</b>) WGA-FITC (green) strongly accumulates in cytoplasmic vesicles of various sizes. Weaker juxtanuclear signals resembling Golgi membranes are also visible (intensity heat map). (<b>B</b>) Overlap of WGA-FITC signal with different cellular compartment markers (red). Higher magnification images of selected areas were recorded with longer exposure times to demonstrate the overlap of WGA-positive vesicular signals with EEA1-positive early endosomes. Cell nuclei were stained with DAPI (blue). Scale bars, 10 µm and 1 µm in higher magnifications.</p>
Full article ">Figure 3
<p>WGA-FITC accumulates at the cVAC in HCMV-infected cells at 120 hpi. Fibroblasts were labeled with WGA-FITC (10 µg/mL) with a 60-min pulse and a 30-min chase, both at either 37 °C or 4 °C. At 37 °C, WGA-FITC (green) is internalized and accumulates as a diffuse signal at the cVAC, which is marked by the viral protein pUL99 (magenta) and delimited by the cis-Golgi marker GM130 (red). In addition, WGA-FITC accumulates in vesicles of different sizes outside the cVAC. Internalization of WGA is inhibited at 4 °C, leading to its accumulation at the plasma membrane. Cell nuclei were stained with DAPI (blue). Scale bars, 10 µm.</p>
Full article ">Figure 4
<p>Characterization of the endocytic compartment in HCMV-infected fibroblasts. Higher magnification images were recorded with longer exposure time. (<b>A</b>) Screening of cellular compartment markers (red) in relation to WGA-FITC (green) and pUL99 (magenta). Fibroblasts were labeled with WGA-FITC (10 µg/mL) with a 60-min pulse and a 30-min chase, both at 37 °C. Higher magnifications show that the punctate WGA-FITC signals at the periphery of the cVAC overlap with EEA1-positive early endosomes and CD63-positive MVBs. The diffuse WGA-FITC signal within the cVAC exhibits some co-localization with γ-Adaptin. (<b>B</b>) Infected fibroblasts were simultaneously labeled with WGA-FITC (10 µg/mL) and Tf-AF555 (50 µg/mL) with a 60-min pulse and a 30-min chase, both at 37 °C. WGA-FITC (green) and Transferrin (Tf, red) localize at the cVAC in a similar pattern. Cellular markers are shown in magenta. Cell nuclei were stained with DAPI (blue). Scale bars, 10 µm and 1 µm in higher magnifications.</p>
Full article ">Figure 5
<p>Localization of WGA relative to nucleocapsids. Infected fibroblasts were labeled with a 24-h pulse of BrdU (10 µM) at 96 hpi, followed by WGA-FITC labeling with a 60-min pulse and a 30-min chase at 120 hpi, both at 37 °C. Labeling of viral DNA by BrdU (red) shows the juxtanuclear accumulation of HCMV nucleocapsids at similar regions as the diffuse WGA-FITC signal (green) at the cVAC, here delimited by the GM130 signal (magenta). Cell nuclei were stained with DAPI (blue). Scale bars, 10 µm.</p>
Full article ">Figure 6
<p>Secondary envelopment at WGA-positive membranes. (<b>A</b>) Overview of an HCMV-infected fibroblast labeled with WGA-HRP. Nu nucleus, Cy cytoplasm, cVAC cytoplasmic viral assembly complex. (<b>B</b>) WGA-positive membrane compartments within the cVAC, including tubular and spherical vesicles (<b>B</b>–<b>E</b>) or Golgi cisternae ((<b>C</b>), white arrowheads). (<b>C</b>–<b>G</b>) Capsids at various stages of secondary envelopment and their association with WGA-positive (black boxes) or WGA-negative (white boxes) vesicles. Enveloped capsids (solid boxes), budding capsids (dashed boxes), naked capsid (black arrowhead), M microtubule organizing center. (<b>F</b>) Capsid budding into a WGA-positive vesicle and (<b>G</b>) enveloped capsid within a WGA-positive vesicle. Quantitative results of this cell in <a href="#app1-biomolecules-14-01149" class="html-app">Figure S5B</a> “cell 7”. Scale bars 5 µm (<b>A</b>,<b>B</b>), 400 nm (<b>C</b>,<b>D</b>,<b>E</b>), 100 nm (<b>F</b>,<b>G</b>).</p>
Full article ">Figure 7
<p>Secondary envelopment of virus particles. Capsid (<b>A</b>) and dense body (DB) (<b>B</b>) bud into WGA-positive membranes, suggesting a common envelopment mechanism. Capsid (<b>C</b>) and DB (<b>D</b>) during secondary envelopment in control samples without WGA-HRP labeling. The membrane profiles show no obvious difference when the membranes were labeled with WGA-HRP. Scale bars, 50 nm.</p>
Full article ">Figure 8
<p>Budding of capsids at WGA-positive membranes as early as 30 min. Infected fibroblast incubated with WGA-HRP for a 10-min pulse at 4 °C and a 30-min chase at 37 °C. (<b>A</b>) Overview. Nu nucleus, Cy cytoplasm, cVAC cytoplasmic viral assembly complex. (<b>B</b>) A region of the cVAC with WGA-positive large endosomes (white asterisks), and capsids. (<b>C</b>) Capsid budding at or (<b>D</b>) capsid enveloped in WGA-positive membrane. (<b>E</b>) Capsid budding at or (<b>F</b>) enveloped in WGA-negative membrane. (<b>G</b>) The trans-site of the Golgi apparatus is occupied by multiple WGA-positive vesicles (arrows). Note the budding event at the vesicle with a striped pattern of DAB precipitate (inset). (<b>H</b>) Trans-most stacked Golgi cisternae also exhibit a striped pattern of DAB precipitate (black asterisks). (<b>I</b>) MVB is filled with numerous WGA-positive intraluminal vesicles. Scale bars, 200 nm.</p>
Full article ">Figure 9
<p>Capsids associated with WGA-positive Golgi membranes. Cells were labeled with WGA-HRP for a 60-min pulse and a 30-min chase. Note the WGA-positive membranes at terminal regions of the trans-sided Golgi cisternae. Scale bar, 500 nm.</p>
Full article ">Figure 10
<p>Three-dimensional (3D) visualization of the Golgi region in an HCMV-infected fibroblast. (<b>A</b>) Stacked Golgi cisternae with the cis-side facing the nucleus and the trans-side facing the cVAC. Capsids and DBs budding into or enveloped by membranes that are not connected to the Golgi cisternae (dark red) and capsids budding into Golgi cisternae were observed. (<b>B</b>–<b>E</b>) Capsids budding at the trans-most Golgi cisterna (red). (<b>D</b>,<b>E</b>) The connection of the budding capsid with the Golgi stack could only be visualized by 3D electron microscopy. Scale bar, 100 nm. Video of this dataset in <a href="#app1-biomolecules-14-01149" class="html-app">Supplementary Movie S1</a>.</p>
Full article ">
20 pages, 3823 KiB  
Article
The Autophagy Receptor SQSTM1/p62 Is a Restriction Factor of HCMV Infection
by Nadine Krämer, Uxía Gestal Mato, Steffi Krauter, Nicole Büscher, Ahmad Afifi, Lina Herhaus, Luise Florin, Bodo Plachter and Christine Zimmermann
Viruses 2024, 16(9), 1440; https://doi.org/10.3390/v16091440 - 10 Sep 2024
Viewed by 420
Abstract
(1) Background: Intrinsic defense mechanisms are pivotal host strategies to restrict viruses already at early stages of their infection. Here, we addressed the question of how the autophagy receptor sequestome 1 (SQSTM1/p62, hereafter referred to as p62) interferes with human cytomegalovirus [...] Read more.
(1) Background: Intrinsic defense mechanisms are pivotal host strategies to restrict viruses already at early stages of their infection. Here, we addressed the question of how the autophagy receptor sequestome 1 (SQSTM1/p62, hereafter referred to as p62) interferes with human cytomegalovirus (HCMV) infection. (2) Methods: CRISPR/Cas9-mediated genome editing, mass spectrometry and the expression of p62 phosphovariants from recombinant HCMVs were used to address the role of p62 during infection. (3) Results: The knockout of p62 resulted in an increased release of HCMV progeny. Mass spectrometry revealed an interaction of p62 with cellular proteins required for nucleocytoplasmic transport. Phosphoproteomics further revealed that p62 is hyperphosphorylated at position S272 in HCMV-infected cells. Phosphorylated p62 showed enhanced nuclear retention, which is concordant with enhanced interaction with viral proteins relevant for genome replication and nuclear capsid egress. This modification led to reduced HCMV progeny release compared to a non-phosphorylated version of p62. (4) Conclusions: p62 is a restriction factor for HCMV replication. The activity of the receptor appears to be regulated by phosphorylation at position S272, leading to enhanced nuclear localization, viral protein degradation and impaired progeny production. Full article
(This article belongs to the Special Issue Molecular Biology of Human Cytomegalovirus)
Show Figures

Figure 1

Figure 1
<p>Impact of p62 and optineurin on HCMV infection. (<b>a</b>,<b>b</b>) Immunoblot analysis of HFF ko-<span class="html-italic">SQSTM1</span> and HFF ko-<span class="html-italic">OPTN</span>, using antibodies against p62, optineurin and GAPDH. (<b>c</b>,<b>d</b>) Quantitative PCR-analysis of HCMV genome replication, following infection of HFF ko-<span class="html-italic">SQSTM1</span> (<b>c</b>) or HFF ko-<span class="html-italic">OPTN</span> (<b>d</b>). Cells were infected with 4 genome copies/cell and collected at the indicated time points post-infection. Genome copies of the isolated viral DNA were determined by TaqMan qPCR. Each value represents the mean of triplicate determinations from three independent experiments. The corresponding standard deviation (SD) is represented by an error bar. (<b>e</b>,<b>f</b>) Quantitation of viral progeny release by the IE1 assay, following infection of HFF ko-<span class="html-italic">SQSTM1</span> (<b>e</b>) or HFF ko-<span class="html-italic">OPTN</span> (<b>f</b>) with HCMV (strain BADwt). The data represent mean values + standard deviations (SD) of eight technical replicates from four (HFF ko-<span class="html-italic">SQSTM1</span>) or three (HFF ko-<span class="html-italic">OPTN</span>) individual experiments for each cell line and the corresponding time point. (<b>g</b>,<b>h</b>) Quantitation of viral progeny release by the IE1 assay, following infection of HFF ko-<span class="html-italic">SQSTM1</span> (<b>g</b>) or HFF ko-<span class="html-italic">OPTN</span> (<b>h</b>) with HCMV (strain TB40). The data represent mean values + standard deviations (SD) of eight technical replicates from three individual experiments for each cell line and the corresponding time point. The statistical analysis was performed by utilizing Welch’s <span class="html-italic">t</span>-test. Not significant (ns): <span class="html-italic">p</span> &gt; 0.05. ***: <span class="html-italic">p</span> ≤ 0.001. ****: <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 1 Cont.
<p>Impact of p62 and optineurin on HCMV infection. (<b>a</b>,<b>b</b>) Immunoblot analysis of HFF ko-<span class="html-italic">SQSTM1</span> and HFF ko-<span class="html-italic">OPTN</span>, using antibodies against p62, optineurin and GAPDH. (<b>c</b>,<b>d</b>) Quantitative PCR-analysis of HCMV genome replication, following infection of HFF ko-<span class="html-italic">SQSTM1</span> (<b>c</b>) or HFF ko-<span class="html-italic">OPTN</span> (<b>d</b>). Cells were infected with 4 genome copies/cell and collected at the indicated time points post-infection. Genome copies of the isolated viral DNA were determined by TaqMan qPCR. Each value represents the mean of triplicate determinations from three independent experiments. The corresponding standard deviation (SD) is represented by an error bar. (<b>e</b>,<b>f</b>) Quantitation of viral progeny release by the IE1 assay, following infection of HFF ko-<span class="html-italic">SQSTM1</span> (<b>e</b>) or HFF ko-<span class="html-italic">OPTN</span> (<b>f</b>) with HCMV (strain BADwt). The data represent mean values + standard deviations (SD) of eight technical replicates from four (HFF ko-<span class="html-italic">SQSTM1</span>) or three (HFF ko-<span class="html-italic">OPTN</span>) individual experiments for each cell line and the corresponding time point. (<b>g</b>,<b>h</b>) Quantitation of viral progeny release by the IE1 assay, following infection of HFF ko-<span class="html-italic">SQSTM1</span> (<b>g</b>) or HFF ko-<span class="html-italic">OPTN</span> (<b>h</b>) with HCMV (strain TB40). The data represent mean values + standard deviations (SD) of eight technical replicates from three individual experiments for each cell line and the corresponding time point. The statistical analysis was performed by utilizing Welch’s <span class="html-italic">t</span>-test. Not significant (ns): <span class="html-italic">p</span> &gt; 0.05. ***: <span class="html-italic">p</span> ≤ 0.001. ****: <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 2
<p>MS analysis of the interaction of p62 (<b>a</b>) and optineurin (<b>b</b>) with HCMV proteins. HFFs, infected with HCMV (m.o.i.= 1) were incubated with protein A/G magnetic beads and specific antibodies against either p62 or optineurin or IgG as control. Precipitates were analyzed and quantified by MS. They were considered significant under the following conditions: one-sided two-sample Student’s <span class="html-italic">t</span>-test with a minimal enrichment factor of 2 (log2(2) = 1), showing the log2 fold change and <span class="html-italic">p</span> &lt; 0.01 (−log10(0.01) = 2). Hits in the upper right quarter are considered significant. All data are deposited at the PRIDE repository (PXD055196).</p>
Full article ">Figure 3
<p>Western blot analysis of viral protein expression in HFF ko-<span class="html-italic">SQSTM1</span>. (<b>a</b>) Viral protein levels in dependence of p62 in HCMV-infected cells. HFF ko-<span class="html-italic">SQSTM1</span> and control cells were infected with HCMV strain TB40, using 8 genome copies/cell. At 1, 2 and 3 d.p.i., cells were collected and subjected to Western blot. The membrane was probed with antibodies against the viral proteins major capsid protein (MCP), pUL57, pUL44, pp28, and pUL48a. GAPDH levels were used as a loading control. (<b>b</b>) ISG levels in dependence of p62 in HCMV-infected cells. HFF ko-<span class="html-italic">SQSTM1</span> and control cells were infected with HCMV, using an m.o.i. of 0.5. At 1, 3 and 6 d.p.i., cells were collected and subjected to Western blot. The membrane was probed with antibodies against the ISGs proteins Mx1 and ISG15. Antibodies against p62 were used as controls. The detection of pp28 served as a control for HCMV infection. GAPDH levels were used as a loading control.</p>
Full article ">Figure 4
<p>Western blot analysis of the influence of p62 and optineurin on autophagy induction. (<b>a</b>) HFF ko-<span class="html-italic">SQSTM1</span>, HFF ko-<span class="html-italic">OPTN</span>, and control cells were infected with HCMV, using an m.o.i. of 0.5. At 1, 3 and 6 d.p.i., cells were collected, and autophagy was analyzed by Western blot. LC3B was used as an indicator of autophagy functions. The turnover of the cytosolic form (LC3B-I) to the autophagosomal membrane associated from (LC3B-II) was detected with an LC3B-specific antibody. Antibodies against p62 and optineurin were applied for control. Detection of the viral IE1 protein at 1 and 3 d.p.i. and pp28 at 6 d.p.i. served as control for HCMV infection. The levels of GAPDH were used as loading control. Shown is a representative Western blot out of four experiments. (<b>b</b>) Ratio of LC3BII to GAPDH from four biological replicates. The statistical difference between each ko-<span class="html-italic">SQSTM1</span> and ko-<span class="html-italic">OPTN</span> was analyzed with two-way ANOVA with Sidak’s multiple comparisons test, ns: not significant.</p>
Full article ">Figure 5
<p>Determination of the phosphorylation status of p62 during HCMV infection. (<b>a</b>) MS analysis of the phosphorylation status of p62 in HCMV-infected versus non-infected HFF-<span class="html-italic">GFP-SQSTM1</span>. The detected phosphorylation sites of p62 of three biological replicates are displayed in a volcano plot, showing the fold change (X-axis) and significance as −log10 <span class="html-italic">p</span>-value (Y-axis). The phosphorylation sites are shown as individual data points. Changes in the phosphorylation status at S272 (colored dot) in infected versus non-infected cells reached significance above the threshold for the fold change of log2 ≥ 1 and <span class="html-italic">p</span>-value-log10 ≤ 0.05, which was represented by the vertical and horizontal dotted lines, respectively. Phosphorylation sites that were detected but did not show significant differences are shown as gray data points. (<b>b</b>) Validation of the phosphoproteomic data in B was performed by Western blot, using a phospho-specific antibody against p62-S272. Lysates of HCMV-infected normal HFF cells were submitted to SDS-PAGE, which was followed by Western blot analysis. The phosphorylation level of p62 at S272 was analyzed at 1, 3, and 6 d.p.i. GAPDH levels were used as loading control. One Western blot of two individual analyses is shown.</p>
Full article ">Figure 6
<p>(<b>a</b>) Construction of different HCMV strains expressing p62-S272 mutants. The BAC technology was used to generate the mutant strains. All recombinant viruses were based on the HCMV parental strain BADwt. The gene region <span class="html-italic">UL1-6</span> of the parental strain was replaced by the <span class="html-italic">SQSTM1</span> gene, encoding the different mutations at the position 272 of SQSTM/p62 [serine (wt), alanine (A), or aspartate (D)]. The BACmids were reconstituted by transfection into HFFs, resulting in the viruses HCMV-p62-S272wt, HCMV-p62-S272A, and HCMV-p62-S272D. The expression of the gene was driven by the modified HCMV major immediate-early promotor (MIEP) with a non-functional cis-repressive sequence (crs) to allow a permanent expression of the respective <span class="html-italic">SQSTM1</span> genes in infected cells. (<b>b</b>,<b>c</b>) Western blot analysis of the p62 levels in ko-<span class="html-italic">SQSTM1</span> cells infected with different HCMV-<span class="html-italic">SQSTM1</span>-S272 strains. (<b>b</b>) Analysis of p62 levels in ko-<span class="html-italic">SQSTM1</span> cells, infected with the different HCMV-<span class="html-italic">SQSTM1</span>-S272 strains (wt/A/D). HCMV-infected wild-type (wt) HFFs were used as a control for p62. Lysates of 5 d.p.i. infected cells were collected and analyzed by Western blot with an antibody directed against p62. Viral pp28 levels were used as infection control, GAPDH levels were used as loading control. One representative Western blot from two analyses is shown. (<b>c</b>) Analysis of the levels of phosphorylation of p62 at position 272, following infection of ko-<span class="html-italic">SQSTM1</span> cells. Cells were infected as in (<b>a</b>) and harvested at 5 d.p.i. Samples were probed for the phosphorylation level of p62 at position 272, using a phospho-specific antibody. A representative Western blot from two analyses is shown.</p>
Full article ">Figure 7
<p>(<b>a</b>–<b>c</b>) Protein–protein interaction networks of the cellular interactors of p62 in dependence on the phosphorylation status at S272, analyzed in samples from co-immunoprecipitation analysis of HCMV-infected HFFs, using the STRING database (<a href="http://string-DBs.org" target="_blank">http://string-DBs.org</a>, accessed on 10 August 2023). HFFs were infected with either HCMV-p62-S272wt, HCMV-p62-S272A, or HCMV-p62-S272D, respectively, using an m.o.i. of 0.5. Cells were collected at three d.p.i. p62 was immunoprecipitated using a receptor-specific antibody. The amount of p62 used for IP was adjusted to control for reduced steady-state levels of HCMV-p62-S272A-infected cells by using twice the number of lysed cells for IP against p62-S272A compared to p62-S272wt or p62-S272D. Each node represents a protein, and the red nodes represent a cluster of proteins, which are associated with the nuclear pore complex or with nucleocytoplasmic transport. The interaction network was generated with a high confidence interaction score (0.7). Networks of proteins that co-precipitated with p62-S272wt are shown in (<b>a</b>), those co-precipitated with p62-S272A are shown in (<b>b</b>), and those co-precipitated with p62-S272D are shown in (<b>c</b>).</p>
Full article ">Figure 8
<p>Indirect immunofluorescence analysis of the localization of p62 in infected cells, depending on S272 phosphorylation. Cells were infected for six days and stained with antibodies against p62 (green) and pp150 (purple). DAPI was used to stain nuclei (blue).</p>
Full article ">Figure 9
<p>Western blot analysis of the impact of S272 phosphorylation on the proteasomal and autolysosomal degradation of p62. (<b>a</b>) Western blot analysis of p62 levels, following 5-day infection of ko-SQSTM1 cells with respective HCMV-p62-S272 strains (wt/D/A) using an m.o.i. of 0.2. Cells were treated with 10 µM of the proteasomal inhibitor MG132 18 h before sample collection. The results of one of two individual experiments are shown. (<b>b</b>) Western blot analysis of p62 levels applying 200 µM of the lysosomal inhibitor bafilomycin A1 (BafA1) 4 h before cell harvest, <span class="html-italic">n</span> = 1. (<b>a</b>,<b>b</b>) Cell lysates were probed with antibodies against p62, GAPDH (loading control), and LC3B.</p>
Full article ">Figure 10
<p>Analysis of HCMV protein expression, genome replication and progeny production in dependence on p62-272 phosphorylation. HFF-ko-SQSTM1 cells were infected with HCMV-p62-S272wt, HCMV-p62-S272A, and HCMV-p62-S272D strains, respectively, using an m.o.i. of 0.1. Cells and cell culture supernatants were collected at the indicated time points. (<b>a</b>) Quantitative PCR analysis of HCMV genome replication. The mean values of three technical replicates from three independent experiments are shown for each virus and time point. The corresponding SD is displayed as error bars. (<b>b</b>) Western blot analysis of the intracellular levels of selected viral proteins, following infection. Cells were harvested at 1, 3, and 6 d.p.i., lysed, and probed by Western blot, using antibodies against HCMV proteins MCP, pp150, pp28 and SCP. The levels of GAPDH were used as loading control. One out of two individual experiments is shown. (<b>c</b>) Viral progeny release, which is measured by the IE1 assay. The graph represents mean values of eight technical replicates from three independent experiments for each virus and time point. The mean values and corresponding SD are represented in a bar chart with error bars. Statistical analysis was performed utilizing Brown–Forsythe and Welch one-way ANOVA (not significant (ns): <span class="html-italic">p</span> &gt; 0.05; *: <span class="html-italic">p</span> &lt; 0.05. ****: <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
10 pages, 538 KiB  
Article
Unveiling the Impact of Human Herpesviruses-Associated on CNS Infections: An Observational Study
by Caio Cesar L. B. Barrionuevo, Pedro P. A. Baptista, Ewerton F. da Silva, Bernardo M. da Silva, Cássia da L. Goulart, Sabrina A. de Melo, Valderjane A. da Silva, Lara Laycia A. de Souza, Rossicleia L. Monte, Fernando F. Almeida-Val, Pablo Vinícius S. Feitoza and Michele de S. Bastos
Viruses 2024, 16(9), 1437; https://doi.org/10.3390/v16091437 - 9 Sep 2024
Viewed by 325
Abstract
Human Herpesviruses (HHVs) play a significant role in neurological diseases such as encephalitis and meningitis, adding significant morbidity. This study aims to retrospectively analyze the effect of HHVs on patients with neurological symptoms, focusing on the Herpesviridae family’s contributions to central nervous system [...] Read more.
Human Herpesviruses (HHVs) play a significant role in neurological diseases such as encephalitis and meningitis, adding significant morbidity. This study aims to retrospectively analyze the effect of HHVs on patients with neurological symptoms, focusing on the Herpesviridae family’s contributions to central nervous system (CNS) infections. Methods: This retrospective cohort study included 895 patients suspected of viral CNS infections, utilizing molecular diagnosis via qPCR to identify HHVs in cerebrospinal fluid (CSF) samples. This was conducted at a reference tertiary care hospital for infectious diseases in the western Brazilian Amazon from January 2015 to December 2022, focusing on the Herpesviridae family’s clinical repercussions and of Cytomegalovirus in CNS infections. Results: The findings revealed that 7.5% of the analyzed samples tested positive for HHVs, with Human Cytomegalovirus (HCMV) and Epstein–Barr Virus (EBV) being the most prevalent. A significant association was found between HHVs and neurological diseases such as encephalitis and meningitis, especially among people living with HIV/AIDS (PLWHA), highlighting the opportunistic nature of these viruses. The study underscores the critical role of CSF analysis in diagnosing CNS infections and the complexity of managing these infections in HIV patients due to their immunocompromised status. Conclusions: The results emphasize the need for comprehensive diagnostic approaches and tailored treatment strategies for CNS infections in immunocompromised individuals. The study calls for ongoing research and advancements in clinical practice to improve patient outcomes facing CNS infections, particularly those caused by HHVs. Full article
(This article belongs to the Special Issue Herpesviruses and Associated Diseases)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the study.</p>
Full article ">
15 pages, 1103 KiB  
Review
Cytomegalovirus Retinitis: Clinical Manifestations, Diagnosis and Treatment
by Jing Zhang, Koju Kamoi, Yuan Zong, Mingming Yang, Yaru Zou, Miki Miyagaki and Kyoko Ohno-Matsui
Viruses 2024, 16(9), 1427; https://doi.org/10.3390/v16091427 - 7 Sep 2024
Viewed by 585
Abstract
Cytomegalovirus (CMV) retinitis is the most common eye disease associated with CMV infection in immunocompromised individuals. The CMVR may initially be asymptomatic; however, relatively mild vitreous inflammation at the onset may be an important differential point from other diseases in HIV patients. Fundus [...] Read more.
Cytomegalovirus (CMV) retinitis is the most common eye disease associated with CMV infection in immunocompromised individuals. The CMVR may initially be asymptomatic; however, relatively mild vitreous inflammation at the onset may be an important differential point from other diseases in HIV patients. Fundus photography, CD4 T-cell count, and telemedicine could be used to screen and monitor the high-risk population, particularly in resource-limited regions. Retinitis generally starts in the peripheral retina and advances toward the posterior pole, which could develop to the characteristic “pizza pie” appearance marked by central retinal necrosis and intraretinal hemorrhage. CMVR causes vision loss if left untreated, and early antiviral therapy significantly reduces the risk of vision loss. Alongside traditional antiviral treatments, immunotherapies including CMV-specific adoptive T-cell therapy and CMV immunoglobulin (CMVIG) are emerging as promising treatment options due to their favorable tolerability and reduced mortality. This review comprehensively examines CMV retinitis, encompassing the clinical features, differential diagnosis, laboratory tests, and updated treatment strategies to inform clinical management. Full article
(This article belongs to the Special Issue Ocular Diseases in Viral Infection)
Show Figures

Figure 1

Figure 1
<p>Fundus photograph of cytomegalovirus retinitis: (<b>A</b>) wedge-shaped appearance with the apex “pointing” toward the optic disc. (<b>B</b>) a “hemorrhagic” appearance involving the posterior pole, characterized by retinal necrosis and edema, intraretinal hemorrhage and “satellite lesions” at the border. (<b>C</b>) cytomegalovirus retinitis involving the periphery, characterized by a “granular” appearance and without hemorrhage. (Modified from Standardization of Uveitis Nomenclature (SUN) Working Group, American journal of ophthalmology, 2021, 228: 245–254 [<a href="#B43-viruses-16-01427" class="html-bibr">43</a>], under a Creative Commons licence CC BY).</p>
Full article ">Figure 2
<p>Cytomegalovirus (CMV) retinitis zones: Zone 1 encompasses the area within 1500 µm of the optic nerve or 3000 µm of the fovea. Zone 2 extends from the outer boundary of Zone 1 to the equator, as determined by the vortex veins. Zone 3 covers the peripheral retina from the equator to the ora serrata (Photo by Kwon H J, et al., Microorganisms, 2021 [<a href="#B48-viruses-16-01427" class="html-bibr">48</a>], under a Creative Commons licence CC BY 4.0).</p>
Full article ">Figure 3
<p>Fundus photographs and OCT images of CMVR and cytomegalovirus papillitis: (<b>A</b>) First visit: fundus photography showed yellowish white retinal necrosis and retinal hemorrhage around the disc; OCT showed exudative retinal detachment in macular area and significant thickening and hyperreflective in temporal retina of optic disc with full-thickness disruption of retinal architecture (yellow arrow). (<b>B</b>) Two weeks: after 2 wk of anti-cytomegalovirus therapy, retinal necrotic lesion has disappeared on fundus photograph, OCT showed subretinal fluid absorption and the edema of the necrotic lesion relief (yellow arrow). (<b>C</b>) Six weeks: fundus photography showed optic atrophy and the retinal necrosis and most of the retinal hemorrhage were absorbed; OCT showed complete absorption of subretinal fluid and retinal thinning in temporal retina of optic disc (yellow arrow). (Photo by Sheng, Yan, et al. International Journal of Ophthalmology, 2020, 13(11): 1800 [<a href="#B77-viruses-16-01427" class="html-bibr">77</a>], under a Creative Commons licence CC BY 4.0).</p>
Full article ">
15 pages, 2516 KiB  
Review
The Effects of Viral Infections on the Molecular and Signaling Pathways Involved in the Development of the PAOs
by Xiaozhou Liu, Zhengdong Zhao, Xinyu Shi, Yanjun Zong and Yu Sun
Viruses 2024, 16(8), 1342; https://doi.org/10.3390/v16081342 - 22 Aug 2024
Viewed by 567
Abstract
Cytomegalovirus infection contributes to 10–30% of congenital hearing loss in children. Vertebrate peripheral auditory organs include the outer, middle, and inner ear. Their development is regulated by multiple signaling pathways. However, most ear diseases due to viral infections are due to congenital infections [...] Read more.
Cytomegalovirus infection contributes to 10–30% of congenital hearing loss in children. Vertebrate peripheral auditory organs include the outer, middle, and inner ear. Their development is regulated by multiple signaling pathways. However, most ear diseases due to viral infections are due to congenital infections and reactivation and affect healthy adults to a lesser extent. This may be due to the fact that viral infections affect signaling pathways that are important for the development of peripheral hearing organs. Therefore, an in-depth understanding of the relationship between viral infections and the signaling pathways involved in the development of peripheral hearing organs is important for the prevention and treatment of ear diseases. In this review, we summarize the effects of viruses on signaling pathways and signaling molecules in the development of peripheral auditory organs. Full article
(This article belongs to the Special Issue 65-Year Anniversary of the Discovery of Cytomegalovirus)
Show Figures

Figure 1

Figure 1
<p>Signals that regulate otic plate (OP) formation. Secretion of FGF8 by the pharyngeal endoderm induces secretion of FGF by the mesoderm near the hindbrain, and FGF defines the posterior PPR (blue) and induces expression of Wnt8a and FGF in the hindbrain. Anterior and posterior hindbrain-derived Wnt8a and FGF-mediated signaling designates the PPR as OP (green).</p>
Full article ">Figure 2
<p>HCMV regulates the classical Wnt/β-catenin signaling pathway. When the Wnt pathway is switched off (the Wnt receptor complex is not bound to the ligand), GSK3α/β phosphorylates β-catenin, which is then ubiquitinated and rapidly destroyed by proteasome targeting. In the nucleus, binding of Groucho to TCF/LEF inhibits transcription of Wnt target genes. When the Wnt signaling pathway is opened, the Fzd/LRP receptor complex activates the classical signaling pathway. Receptor recruitment to Dvl serves as a docking platform for components of the β-catenin disruption complex. Wnt induces GSK3β and CK1/CK2 to phosphorylate LRP5/6, which regulates Axin docking and releases β-catenin. In the nucleus, β-catenin replaces Groucho in Tcf/Lef, thereby promoting transcription of Wnt target genes. HCMV reduces the poly-ADP-ribosylation activity of end-anchor polymerase, thereby stabilizing Axin and inhibiting the Wnt pathway. HCMV US28 inhibits GSK3 activity via the ROCK pathway, β-catenin cannot be phosphorylated for degradation HCMV US28 inhibits GSK3 activity through the ROCK pathway, and β-catenin cannot be phosphorylated and degraded.</p>
Full article ">Figure 3
<p>HCMV regulates the Notch signaling pathway. When Notch receptor binds to Notch ligand, Notch receptor is cleaved by ADAM10 and γ-secretase, leading to the release of Notch intracellular structural domain (NICD). NICD fragment is the active form of the receptor that binds to the CSL transcription factor to trigger the transcription of the Notch target genes. Hes proteins also promote the phosphorylation and activation of STAT3, and the activated STAT3 shuttles to the nucleus to bind the Sox2 promoter and induce its expression. HCMV is a membrane protein that regulates Notch signaling pathway phosphorylation and activation and activated STAT3 shuttles to the nucleus to bind the Sox2 promoter and induce its expression. HCMV periplasmic protein pp71 can alter the cellular localization of Jag1 and NICD and inhibit Notch signaling by promoting their degradation via the proteasome. HCMV IE1 promotes proteasomal degradation of Hes1 to inhibit the phosphorylation of STAT3 and promotes the accumulation of unphosphorylated STAT3 in the nucleus and inhibits Sox2 transcription.</p>
Full article ">
9 pages, 1077 KiB  
Case Report
Cytomegalovirus, a “Friend” of SARS-CoV-2: A Case Report
by Nicoleta-Ana Tomşa, Lorena Elena Meliţ, Gabriela Bucur, Anca-Meda Văsieșiu and Cristina Oana Mărginean
Children 2024, 11(8), 1010; https://doi.org/10.3390/children11081010 - 19 Aug 2024
Viewed by 577
Abstract
Introduction: Cytomegalovirus (CMV) infection is present in a latent state in 70–90% of the immunocompetent population, and its reactivation might be triggered by inflammatory conditions such as post-COVID multisystem inflammatory syndrome (MIS-C) or by immunosuppression induced by steroids. The aim of this paper [...] Read more.
Introduction: Cytomegalovirus (CMV) infection is present in a latent state in 70–90% of the immunocompetent population, and its reactivation might be triggered by inflammatory conditions such as post-COVID multisystem inflammatory syndrome (MIS-C) or by immunosuppression induced by steroids. The aim of this paper was to highlight the unexpected complications associated with SARS-CoV-2 infection that require a complex clinical approach for accurate diagnosis. Materials and Methods: We present the case of a 4-year-old male patient who, during an initially favorable course of PIMS, experienced symptoms of respiratory failure. Results: The patient initially presented with clinical and paraclinical signs of PIMS with cardiac involvement, for which high-dose corticosteroid therapy was initiated, followed by gradual tapering, along with immunoglobulins, anticoagulants, antiplatelet agents, and symptomatic treatment. After 10 days of favorable progress, the patient’s general condition deteriorated, showing tachypnea, desaturation, and a ground-glass appearance on thoracic CT. Negative inflammatory markers and favorable cardiac lesion evolution ruled out MIS-C relapse. The presence of anti-CMV IgM antibodies and viral DNA in the blood confirmed acute CMV infection, likely triggered by prior severe-acute-respiratory-syndrome-related coronavirus 2 (SARS-CoV-2) infection and secondary immunosuppression due to steroids. Non-specific immunomodulatory treatment was initiated but led to worsening of pulmonary lesions, prompting the initiation of specific antiviral treatment with ganciclovir, resulting in rapid clinical and imaging improvement. Conclusions: CMV infection can be reactivated by immunosuppression induced by corticosteroid therapy for MIS-C and may require specific etiological treatment. Full article
(This article belongs to the Section Pediatric Allergy and Immunology)
Show Figures

Figure 1

Figure 1
<p>Diffuse peribronchovascular infiltrates and inferior interclavicular hilar infiltrates (white arrows).</p>
Full article ">Figure 2
<p>Ground-glass opacities with subpleural disposition adjacent to the mediastinum, in the upper lobes at the apical and posterior segments of the right lung, and at the apicoposterior segment of the lung (white arrows).</p>
Full article ">Figure 3
<p>In the lower pulmonary lobes, opacities were observed in the bilateral posterior segments: left anteromedial and right medial segments (white arrows).</p>
Full article ">
17 pages, 707 KiB  
Review
Burden of Congenital CMV Infection: A Narrative Review and Implications for Public Health Interventions
by Cecilia Liberati, Giulia Sturniolo, Giulia Brigadoi, Silvia Cavinato, Silvia Visentin, Erich Cosmi, Daniele Donà and Osvalda Rampon
Viruses 2024, 16(8), 1311; https://doi.org/10.3390/v16081311 - 17 Aug 2024
Viewed by 480
Abstract
Cytomegalovirus causes the most common congenital infection worldwide. With most infants asymptomatic at birth, the few affected may present with variable clinical scenarios, from isolated hearing loss to severe neurologic impairment. Public health interventions include all actions at the health system, community, and [...] Read more.
Cytomegalovirus causes the most common congenital infection worldwide. With most infants asymptomatic at birth, the few affected may present with variable clinical scenarios, from isolated hearing loss to severe neurologic impairment. Public health interventions include all actions at the health system, community, and individual levels that aim at reducing the burden of congenital Cytomegalovirus. This review examines the literature on maternal and neonatal screening programs in light of current evidence for treatment and the development of vaccines against Cytomegalovirus. Potential biases and benefits of these interventions are outlined, with the objective of increasing awareness about the problem and providing readers with data and critical tools to participate in this ongoing debate. Full article
(This article belongs to the Special Issue 65-Year Anniversary of the Discovery of Cytomegalovirus)
Show Figures

Figure 1

Figure 1
<p>Preventive public measures proposed to reduce the cCMV burden. Adapted from Public Health Interventions (population-based), Minnesota Department of Health [<a href="#B18-viruses-16-01311" class="html-bibr">18</a>].</p>
Full article ">
7 pages, 1215 KiB  
Case Report
From Pancytopenia to Hyperleukocytosis, an Unexpected Presentation of Immune Reconstitution Inflammatory Syndrome in an Infant with Methylmalonic Acidemia
by Samuel Sassine, Amandine Remy, Tanguy Demaret, François Proulx, Julie Autmizguine, Fatima Kakkar, Thai Hoa Tran, Caroline Laverdière, Ellery T. Cunan, Catalina Maftei, Grant Mitchell, Hélène Decaluwe and Jade Hindié
Children 2024, 11(8), 990; https://doi.org/10.3390/children11080990 - 14 Aug 2024
Viewed by 976
Abstract
A 2.5-month-old girl admitted for failure to thrive and severe pancytopenia was diagnosed with methylmalonic acidemia (MMA) secondary to transcobalamin II deficiency, an inborn error of vitamin B12 metabolism. Opportunistic Cytomegalovirus and Pneumocystis jirovecii pneumonia led to severe acute respiratory distress syndrome (ARDS) [...] Read more.
A 2.5-month-old girl admitted for failure to thrive and severe pancytopenia was diagnosed with methylmalonic acidemia (MMA) secondary to transcobalamin II deficiency, an inborn error of vitamin B12 metabolism. Opportunistic Cytomegalovirus and Pneumocystis jirovecii pneumonia led to severe acute respiratory distress syndrome (ARDS) and immune reconstitution inflammatory syndrome (IRIS) after treatment initiation with vitamin B12 supplementation. In children with interstitial pneumonia-related ARDS, normal lymphocyte count should not delay invasive procedures required to document opportunistic infections. MMA can be associated with underlying lymphocyte dysfunction and vitamin B12 supplementation can fully reverse the associated immunodeficiency. IRIS may appear in highly treatment-responsive forms of pancytopenia in children and prompt treatment of dysregulated inflammation with high-dose corticosteroids should be initiated. Full article
(This article belongs to the Section Pediatric Allergy and Immunology)
Show Figures

Figure 1

Figure 1
<p>After hydroxocobalamin treatment, the white blood cell (WBC) count dramatically increased and the respiratory status of the patient deteriorated, both compatible with immune reconstitution inflammatory syndrome (IRIS). One month after hydroxocobalamin treatment, coupled with a 2 week corticosteroid course, complete blood count normalized. White blood cells (<b>A</b>) and neutrophils (<b>B</b>) responded dramatically to hydroxocobalamin treatment. Platelets (<b>C</b>) normalized after a delayed overshoot. Hemoglobin (<b>D</b>) slowly normalized after a rapid increase in reticulocytes (<b>D</b>) (Rtc, gray curve) soon after hydroxocobalamin injection.</p>
Full article ">Figure 2
<p>Patients X-ray, CT-scan and growth charts. Chest CT-scan confirming severe interstitial lung disease (<b>A</b>), chest X-ray showing interstitial lung disease (<b>B</b>), patient weight (<b>C</b>) and height (<b>D</b>) growth chart. The red lines represent the patient’s curve.</p>
Full article ">
32 pages, 4717 KiB  
Review
Understanding the Cytomegalovirus Cyclin-Dependent Kinase Ortholog pUL97 as a Multifaceted Regulator and an Antiviral Drug Target
by Manfred Marschall, Martin Schütz, Markus Wild, Eileen Socher, Christina Wangen, Kishore Dhotre, William D. Rawlinson and Heinrich Sticht
Cells 2024, 13(16), 1338; https://doi.org/10.3390/cells13161338 - 13 Aug 2024
Viewed by 1048
Abstract
Herpesviral protein kinases, such as the therapy-relevant pUL97 of human cytomegalovirus (HCMV), are important for viral replication efficiency as well as pathogenesis, and represent key antiviral drug targets. HCMV pUL97 is a viral cyclin-dependent kinase (CDK) ortholog, as it shares functional and structural [...] Read more.
Herpesviral protein kinases, such as the therapy-relevant pUL97 of human cytomegalovirus (HCMV), are important for viral replication efficiency as well as pathogenesis, and represent key antiviral drug targets. HCMV pUL97 is a viral cyclin-dependent kinase (CDK) ortholog, as it shares functional and structural properties with human CDKs. Recently, the formation of vCDK/pUL97–cyclin complexes and the phosphorylation of a variety of viral and cellular substrate proteins has been demonstrated. Genetic mapping and structural modeling approaches helped to define two pUL97 interfaces, IF1 and IF2, responsible for cyclin binding. In particular, the regulatory importance of interactions between vCDK/pUL97 and host cyclins as well as CDKs has been highlighted, both as determinants of virus replication and as a novel drug-targeting option. This aspect was substantiated by the finding that virus replication was impaired upon cyclin type H knock-down, and that such host-directed interference also affected viruses resistant to existing therapies. Beyond the formation of binary interactive complexes, a ternary pUL97–cyclin H–CDK7 complex has also been described, and in light of this, an experimental trans-stimulation of CDK7 activity by pUL97 appeared crucial for virus–host coregulation. In accordance with this understanding, several novel antiviral targeting options have emerged. These include kinase inhibitors directed to pUL97, to host CDKs, and to the pUL97–cyclin H interactive complexes. Importantly, a statistically significant drug synergy has recently been reported for antiviral treatment schemes using combinations of pharmacologically relevant CDK7 and vCDK/pUL97 inhibitors, including maribavir. Combined, such findings provide increased options for anti-HCMV control. This review focuses on regulatory interactions of vCDK/pUL97 with the host cyclin–CDK apparatus, and it addresses the functional relevance of these key effector complexes for viral replication and pathogenesis. On this basis, novel strategies of antiviral drug targeting are defined. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the HCMV-encoded protein kinase pUL97 and its various substrates. The CDK-like protein kinase pUL97 of human cytomegalovirus interacts with cyclins and phosphorylates (P), as well as a number of viral (encircled in orange) and cellular (encircled in green) substrate proteins. Functional assignment of the individual proteins is indicated by curly brackets. RNAPII, cellular RNA polymerase II; EF-1δ, translational elongation factor 1δ; IFI16, interferon-inducible protein 16; SAMHD1, SAM domain and HD domain-containing protein 1; pUL69, pp65, pUL44, pUL50, pUL53, cytomegaloviral early proteins with specific regulatory functions; p32/gC1qR, acidic 32-kDa multiligand-binding protein as a receptor for globular head domain of complement C1q; lamin A/C, human nuclear lamin protein of types A and C; Rb, human retinoblastoma protein.</p>
Full article ">Figure 2
<p>Cyclin interaction sites and model of pUL97–cyclin H complex. (<b>A</b>) Structure of the ternary CDK7–cyclin H–MAT1 complex (PDB code: 7B5O [<a href="#B106-cells-13-01338" class="html-bibr">106</a>]). (<b>B</b>) Structure of the quaternary CDK9–cyclin T–Tat–AFF4 complex (PDB code: 4OR5 [<a href="#B107-cells-13-01338" class="html-bibr">107</a>]). (<b>C</b>) Model of a ternary pUL97–cyclin H–CDK7 complex, in which pUL97 is attached to cyclin H exclusively through IF2 formed by the 231–280 sequence stretch. The pUL97 kinase domain (residues 329–634, marked in red) is connected to the complex by a nonstructured, flexible linker (residues 281–328, indicated as dark orange connecting line). (<b>D</b>) Model of a pUL97–cyclin H complex, in which pUL97 interacts with cyclin H both through IF2 (orange), pUL97(231–280), and the globular kinase domain IF1 (red), pUL97(329–634), thereby displacing CDK7. Panels (<b>C</b>,<b>D</b>) adapted from [<a href="#B105-cells-13-01338" class="html-bibr">105</a>].</p>
Full article ">Figure 3
<p>Importance of pUL97–cyclin H interaction for viral replication and kinase activity. (<b>A</b>) HFFs were infected with HCMV (recombinant TB40-IE2-YFP) WT or pUL97 IF2 deletion mutants. Viral genome equivalents of released virions were determined by qPCR. (<b>B</b>) HFFs infected with HCMV (recombinant TB40-IE2-YFP) WT or pUL97 IF2 deletion mutants at MOI 0.01 were harvested and lysed 4 d.p.i., before cyclin H was immunoprecipitated and the coimmunoprecipitation of pUL97 was analyzed by Wb. (<b>C</b>) Cyclin H protein quantities were analyzed from TEV-1 cells infected with HCMV Merlin by densitometric quantitation of Wbs at specified time points. (<b>D</b>) HFFs with doxycycline-inducible cyclin H knockdown were infected with HCMV AD169, and viral genome equivalents were determined by qPCR (for various cellular controls of this approach, see [<a href="#B102-cells-13-01338" class="html-bibr">102</a>,<a href="#B105-cells-13-01338" class="html-bibr">105</a>,<a href="#B138-cells-13-01338" class="html-bibr">138</a>]). (<b>E</b>) 293T cells transfected with pUL97-Flag or empty vector were analyzed for kinase activity using a qSox-IVKA with increasing concentrations of recombinant cyclin H. (<b>F</b>) pUL97-Flag WT and Δ231–280 mutant were analyzed for kinase activity using a qSox-IVKA. Values were normalized to pUL97-Flag WT. (<b>G</b>) 293T cells transfected with pUL97-Flag or empty vector, and pUL97-Flag and/or CDK7 were immunoprecipitated and analyzed by a qSox-IVKA. Kinase signals were normalized to CDK7. ****, <span class="html-italic">p</span> &lt; 0.0001; ***, <span class="html-italic">p</span> &lt; 0.001; **, <span class="html-italic">p</span> &lt; 0.01; n.s., not significant. For detailed methodology, see [<a href="#B104-cells-13-01338" class="html-bibr">104</a>,<a href="#B139-cells-13-01338" class="html-bibr">139</a>].</p>
Full article ">Figure 4
<p>Scheme summarizing the complex regulatory interaction between vCDK/pUL97, cyclin H, and CDK7. Details of regulatory protein complex formation as well as the aspired drug-targeting functions are depicted. A specific focus is given to the options of developing mechanistically novel antiviral drugs directed to this complex. Detailed explanations are given in <a href="#sec3dot4-cells-13-01338" class="html-sec">Section 3.4</a>, <a href="#sec5dot3-cells-13-01338" class="html-sec">Section 5.3</a>, <a href="#sec6-cells-13-01338" class="html-sec">Section 6</a> and <a href="#sec7-cells-13-01338" class="html-sec">Section 7</a>; for further details, see [<a href="#B105-cells-13-01338" class="html-bibr">105</a>].</p>
Full article ">Figure 5
<p>Schematic depiction of the HCMV replication cycle. Specific consideration is given to approved, direct-acting antivirals and their viral target proteins (light green boxes), as well as experimental host-directed antivirals and their cellular target proteins (dark green boxes). Steps of the viral replication cycle are given in italics. Details of the scheme are explained in the text. The primary data presented in this section, i.e., here appearing in a refined presentation style, have been published elsewhere [<a href="#B141-cells-13-01338" class="html-bibr">141</a>,<a href="#B159-cells-13-01338" class="html-bibr">159</a>].</p>
Full article ">Figure 6
<p>Characterization of the selective CDK7 inhibitor LDC4297 for anti-HCMV activity. (<b>A</b>) Structural formula of LDC4297. (<b>B</b>) Residual activity of top seven most inhibited cellular kinases under treatment with 100 nM LDC4297. Radiometric protein kinase assays were performed for 333 individual protein kinases [<a href="#B140-cells-13-01338" class="html-bibr">140</a>]. Note the low degree of secondary inhibitory activity against kinases other than CDK7; the tumor-relevant mutant of MET kinase, Y1230A, has no relevance for a normal cellular background. (<b>C</b>) Antiviral profile of LDC4297 against HCMV AD169-GFP replication, at MOI of approx. 0.01, as determined by GFP-based replication assay for 7 days, using total lysates of infected HFFs. Data is shown as mean between two biological replicates with standard deviation (SD), normalized to solvent control DMSO (GFP fluorometric measurements in quadruplicate). Half-maximal effective dose (EC<sub>50</sub>) is given in the grey box as mean between two biological replicates ± SD. (<b>D</b>) Cytotoxicity profile of LDC4297 in HFFs as determined by Neutral Red uptake assay. Data is shown as mean between three biological replicates with SD, normalized to solvent control DMSO. Approximation of half-maximal cytotoxic dose (CC<sub>50</sub>) is given in the grey box. Staurosporine (STP) was used as a positive control.</p>
Full article ">Figure 7
<p>In vivo anti-MCMV activity of abemaciclib. (<b>A</b>) Exemplary images of in vivo luciferase imaging of four mice representative for respective treatment group. Color grading representing radiance is given in scale below. (<b>B</b>) Antiviral efficacy of abemaciclib treatment as assessed by in vivo luciferase imaging of spleen and liver. Data is shown as mean between analyzed organs with SD, normalized to vehicle control. (<b>C</b>) Antiviral efficacy of abemaciclib treatment as assessed by luciferase assay in lung and liver homogenates. Data are shown as mean between analyzed organs with SD, normalized to vehicle control. (<b>D</b>) Antiviral efficacy of abemaciclib treatment as assessed by viral genome-specific qPCR of DNA extracted from spleen and liver samples. Data are shown as mean between analyzed organs with SD, normalized to vehicle control. (Note, that the differences in (<b>B</b>–<b>D</b>) represent experimental tendencies that so far lacked statistical significance.</p>
Full article ">Figure 8
<p>Loewe additivity fixed-dose dilution matrix and result visualization. (<b>A</b>) Dilution matrix employed for standard Loewe additivity fixed-dose assay. (<b>B</b>) CI value graph depicting exemplary synergistic drug interaction. Threshold between antagonistic and synergistic interaction (i.e., additive interaction) is marked by green dotted line. CI values at 50%, 75%, 90%, and 95% are given in bold. CI<sub>wt</sub> is given in grey box.</p>
Full article ">Figure 9
<p>Loewe additivity fixed-dose dilution matrix and result visualization. (<b>A</b>) Dilution matrix employed for standard Loewe additivity fixed-dose assay. (<b>B</b>) CI value graph depicting exemplary synergistic interaction of two arbitrary drugs (hypothetical data derived from another context [<a href="#B159-cells-13-01338" class="html-bibr">159</a>]). Threshold between antagonistic and synergistic interaction (i.e., additive interaction) is marked by a green dotted line. CI values at 50%, 75%, 90%, and 95% are given in bold. CI<sub>wt</sub> is given in grey box.</p>
Full article ">Figure 10
<p>Comparative alignment of Bliss independence checkerboard and Loewe additivity fixed-dose results. X-axis value indicates mean synergy volume sum across checkerboard assays. Y-axis value indicates mean CI<sub>wt</sub> across fixed-dose assays. Colored bars represent ranges of antagonistic (blue), and synergistic (green) values for each method. Solid blue/green fields within the chart indicate overlapping antagonistic/synergistic range of both approaches, respectively; dashed blue/green areas designate antagonistic or synergistic ranges in one method with additive values in the other method.</p>
Full article ">Figure 11
<p>Structural depiction of drug-exposed contact amino acids. The comparative modeling setting illustrates the contact residues of vCDK/pUL97 (modeled with AlphaFold) and CDK7 (PDB ID: 8P7L [<a href="#B168-cells-13-01338" class="html-bibr">168</a>]) that are involved in drug binding of MBV or LDC4297, respectively. (<b>A</b>) Prediction of the vCDK/pUL97 kinase domain and MBV docking to its ATP-binding pocket; and in comparison (<b>B</b>), prediction of the CDK7 kinase domain and LDC4297 docking to its ATP-binding pocket.</p>
Full article ">
17 pages, 1746 KiB  
Article
Assessment of Dried Serum Spots (DSS) and Volumetric-Absorptive Microsampling (VAMS) Techniques in Therapeutic Drug Monitoring of (Val)Ganciclovir—Comparative Study in Analytical and Clinical Practice
by Arkadiusz Kocur, Agnieszka Czajkowska, Mateusz Moczulski, Bartłomiej Kot, Jacek Rubik and Tomasz Pawiński
Int. J. Mol. Sci. 2024, 25(16), 8760; https://doi.org/10.3390/ijms25168760 - 12 Aug 2024
Viewed by 524
Abstract
Ganciclovir (GCV) and its prodrug valganciclovir (VGCV) are antiviral medications primarily used to treat infections caused by cytomegalovirus (CMV), particularly in immunocompromised individuals such as solid organ transplant (SOT) recipients. Therapy with GCV is associated with significant side effects, including bone marrow suppression. [...] Read more.
Ganciclovir (GCV) and its prodrug valganciclovir (VGCV) are antiviral medications primarily used to treat infections caused by cytomegalovirus (CMV), particularly in immunocompromised individuals such as solid organ transplant (SOT) recipients. Therapy with GCV is associated with significant side effects, including bone marrow suppression. Therefore, therapeutic drug monitoring (TDM) is mandatory for an appropriate balance between subtherapeutic and toxic drug levels. This study aimed to develop and validate three novel methods based on liquid chromatography-tandem mass spectrometry (LC-MS/MS) for GCV determination in serum (reference methodology), dried serum spots (DSS), and VAMS-Mitra™ devices. The methods were optimized and validated in the 0.1–25 mg/L calibration range. The obtained results fulfilled the EMA acceptance criteria for bioanalytical method validation. Assessment of DSS and VAMS techniques extended GCV stability to serum for up to a minimum of 49 days (at room temperature, with desiccant). Developed methods were effectively evaluated using 80 clinical serum samples from pediatric renal transplant recipients. Obtained samples were used for DSS, and dried serum VAMS samples were manually generated in the laboratory. The results of GCV determination using serum-, DSS- and VAMS-LC-MS/MS methods were compared using regression analysis and bias evaluation. The conducted statistical analysis confirmed the interchangeability between developed assays. The DSS and VAMS samples are more accessible and stable during storage, transport and shipment than classic serum samples. Full article
Show Figures

Figure 1

Figure 1
<p>Molecular mechanisms of (val)ganciclovir action in CMV-infected cells. Mutations in UL97 and UL54 are responsible for GCV resistance. GCV—ganciclovir, VGCV—valganciclovir, GUK1—guanylate kinase 1, NUDT15—nudix hydrolase 15, dNTP—deoxynucleotide triphosphate. Created using bioRender.com (accessed on 8 August 2024).</p>
Full article ">Figure 2
<p>Representative chromatograms of GCV (retention time = 0.63 min): (<b>a</b>) blank sample; (<b>b</b>) serum patient sample—8.66 mg/L GCV concentration; (<b>c</b>) dried serum spot patient sample—8.81 mg/L GCV concentration; (<b>d</b>) s-VAMS patient sample—8.33 mg/L GCV concentration.</p>
Full article ">Figure 3
<p>Results of GCV determination using chromatographic methods expressed as bar/dotted graph with whiskers. Methods are marked as follows: red dots (serum-LC-MS/MS with GCV-d<sub>5</sub> as internal standard), pink dots (serum-LC-MS/MS with ACV as internal standard), green dots (dried serum spots, DSS-LC-MS/MS with ACV as internal standard), blue dots (dried serum in VAMS-LC-MS/MS with ACV as internal standard), green dots (serum-HPLC/UV with AVC as internal standard). GCV—ganciclovir.</p>
Full article ">
13 pages, 2785 KiB  
Article
Targeting Peptidylarginine Deiminase 3 to Efficiently Suppress Herpes Simplex Virus Type 2 Infection
by Selina Pasquero, Francesca Gugliesi, Matteo Biolatti, Camilla Albano, Greta Bajetto, Linda Trifirò, Stefano Raviola, Valentina Dell’Oste and Marco De Andrea
Int. J. Mol. Sci. 2024, 25(16), 8709; https://doi.org/10.3390/ijms25168709 - 9 Aug 2024
Viewed by 764
Abstract
Protein expression is regulated through multiple mechanisms, including post-translational modifications (PTMs), which can alter protein structure, stability, localization, and function. Among these, citrullination stands out due to its ability to convert arginine residues into citrulline, altering protein charge and mass. This modification is [...] Read more.
Protein expression is regulated through multiple mechanisms, including post-translational modifications (PTMs), which can alter protein structure, stability, localization, and function. Among these, citrullination stands out due to its ability to convert arginine residues into citrulline, altering protein charge and mass. This modification is catalyzed by calcium-dependent protein arginine deiminases (PADs), enzymes implicated in various inflammatory diseases. We have recently shown that human cytomegalovirus (HCMV) and herpes simplex virus type 1 (HSV-1) exploit these enzymes to enhance their replication capabilities. Although the role of PADs in HCMV and HSV-1 infections is well documented, their involvement in HSV-2 infection has not yet been thoroughly investigated. Here, we demonstrate that HSV-2 manipulates the overall protein citrullination profile by activating three PAD isoforms: PAD2, PAD3, and PAD4. However, as previously observed during HSV-1 infection, PAD3 is the most significantly upregulated isoform, both at the mRNA and protein levels. Consistently, we demonstrate that inhibiting PAD3, either through the specific inhibitor CAY10727 or via CRISPR/Cas9-mediated gene silencing, markedly reduces HSV-2 replication and viral protein expression. Lastly, we show that CAY10727 displays an IC50 value of 0.3 μM, which is extremely close to what was previously observed for HSV-1. Overall, our findings highlight the crucial role of PAD3 in the life cycle of HSV-2 and suggest that the targeted inhibition of PAD3 may represent a promising approach for treating HSV-2 infections, especially in cases resistant to existing antiviral therapies. Full article
(This article belongs to the Special Issue Recent Advances in Herpesviruses)
Show Figures

Figure 1

Figure 1
<p>HSV-2 infection affects the protein citrullination pattern in HFFs. (<b>A</b>) Schematic representation of the method used for labeling citrullinated proteins in cellular protein lysates using the Rh-PG probe. Created with BioRender.com. (<b>B</b>) Protein lysates from mock-infected or HSV-2-infected HFFs, collected at different hpi, were treated with the Rh-PG probe and analyzed through gel electrophoresis to identify citrullinated proteins. An anti-ICP8 antibody confirmed HSV-2 infection, while β-actin cellular expression served as the protein loading control. One representative blot of three independent experiments is shown. (<b>C</b>) Comparison of total citrullinated protein levels detected in mock- vs. HSV-2-infected HFFs using the Rh-PG probe. Values are expressed as means ± SEM of four independent experiments, * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001; one-way ANOVA followed by Bonferroni’s post-test.</p>
Full article ">Figure 2
<p>HSV-2 infection increases PADI3 expression in human fibroblasts. (<b>A</b>) mRNA expression levels of PADI isoforms by RT-qPCR of HSV-2-infected vs. uninfected (mock) HFFs, normalized to GAPDH and expressed as mean fold change ± SEM over mock-infected cells. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001; one-way ANOVA followed by Bonferroni’s post-test. (<b>B</b>) Protein lysates from uninfected (mock) or infected HFFs were analyzed by immunoblotting at different time points using the indicated antibodies. A representative blot from three independent experiments is presented.</p>
Full article ">Figure 3
<p>Targeting PAD3 hampers HSV-2 replication in human cells. (<b>A</b>) Representative images of infected HFFs (24 hpi) at an MOI of 1 PFU/cell treated with various PAD inhibitors or vehicle (DMSO) (syncytia formation = black arrows). Scale bars: 20 μm. (<b>B</b>) Plaque assay on supernatant of HSV-2-infected HFFs (MOI 1) collected at 24 hpi and treated with BB-Cl-amidine (2.5 μM), AFM30a (20 μM), CAY10727 (1 μM), GSK199 (20 μM), or vehicle. Values are shown as means ± SEM (error bars) from four independent experiments, * <span class="html-italic">p</span> &lt; 0.05; non-parametric paired <span class="html-italic">t</span>-test. (<b>C</b>) Protein lysates from uninfected (mock) or infected HFFs (24 hpi) at an MOI of 1 PFU/cell treated as in (<b>B</b>) were analyzed by Western blot to assess viral expression with anti-ICP8 and anti-gD antibodies; β-actin was employed as a loading control for cellular protein expression. One representative blot of three independent experiments is shown.</p>
Full article ">Figure 4
<p>Antiviral activity and IC50 calculation of PAD inhibitors. HFFs were infected with HSV-2 (MOI 1 PFU/cell) and subsequently treated with the indicated concentrations of BB-Cl (<b>A</b>) or CAY10727 (<b>B</b>). Treatments were administered 1 h before virus adsorption and maintained throughout the experiment. At 24 hpi, viral plaques were counted under a microscope, and the number of plaques are plotted against the inhibitor concentration. Values are presented as means ± SEM (error bars) of four independent experiments. Values are expressed as means ± SEM (error bars) from four independent experiments, * <span class="html-italic">p</span> &lt; 0.05; one-way ANOVA followed by Dunnett’s post-tests. (<b>C</b>,<b>D</b>) Protein lysates from uninfected (mock) or infected HFFs (24 hpi) at an MOI of 1 PFU/cell treated as in (<b>A</b>,<b>B</b>) were subjected to immunoblotting to assess viral expression using anti-ICP8 and anti-gD antibodies; β-actin served as a control for equal loading. One representative blot of three independent experiments is shown. (<b>E</b>) Protein lysates from uninfected (mock) or infected HFFs (24 hpi) at an MOI of 1 PFU/cell treated with CAY10727 (0.5 µM), BB-Cl (2.5 µM), or vehicle (DMSO or Et-OH) were labeled with the Rh-PG probe and subjected to gel electrophoresis to detect citrullinated proteins. (<b>F</b>) Comparison of total citrullinated protein levels detected in (<b>E</b>). Values are presented as means ± SEM of three independent experiments, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001; one-way ANOVA followed by Bonferroni’s post-test.</p>
Full article ">Figure 5
<p>Genetic ablation of PADI3 gene impairs HSV-2 replication. PAD3 knockout (PAD3KO) HFFs were created using CRISPR/Cas9 technology. (<b>A</b>) The effectiveness of PAD3 protein depletion at 24 hpi was evaluated by Western blot with antibodies against PAD3 or β-actin, with the latter serving as the protein loading control. Relative densitometric analysis, representative of three independent experiments, presents values expressed as fold change in PAD3 expression normalized to β-actin. (<b>B</b>) PAD3KO HFFs were infected with HSV-2 (MOI 1). At 24 hpi, the viral supernatants were collected and quantified by the standard plaque assay. Values are expressed as mean ± SEM relative to three independent experiments, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 non-parametric <span class="html-italic">t</span>-test. (<b>C</b>) Western blot analysis on protein lysates from PAD3WT (ctrl) or PAD3KO HFFs infected with HSV-2 (MOI 1) for 24 h. Viral protein expression was determined using the anti-gD antibody, with β-actin expression used to verify equal protein loading. Relative densitometric analysis, representative of three independent experiments, shows values expressed as fold change in viral gD expression normalized to β-actin.</p>
Full article ">
15 pages, 1588 KiB  
Review
No Time to Die: How Cytomegaloviruses Suppress Apoptosis, Necroptosis, and Pyroptosis
by Yingqi Deng, Ana Águeda-Pinto and Wolfram Brune
Viruses 2024, 16(8), 1272; https://doi.org/10.3390/v16081272 - 9 Aug 2024
Viewed by 701
Abstract
Viruses are obligate intracellular pathogens as their replication depends on the metabolism of the host cell. The induction of cellular suicide, known as programmed cell death (PCD), has the potential to hinder viral replication and act as a first line of defense against [...] Read more.
Viruses are obligate intracellular pathogens as their replication depends on the metabolism of the host cell. The induction of cellular suicide, known as programmed cell death (PCD), has the potential to hinder viral replication and act as a first line of defense against viral pathogens. Apoptosis, necroptosis, and pyroptosis are three important PCD modalities. Different signaling pathways are involved in their execution, and they also differ in their ability to cause inflammation. Cytomegaloviruses (CMV), beta-herpesviruses with large double-stranded DNA genomes, encode a great variety of immune evasion genes, including several cell death suppressors. While CMV inhibitors of apoptosis and necroptosis have been known and studied for years, the first pyroptosis inhibitor has been identified and characterized only recently. Here, we describe how human and murine CMV interfere with apoptosis, necroptosis, and pyroptosis signaling pathways. We also discuss the importance of the different PCD forms and their viral inhibitors for the containment of viral replication and spread in vivo. Full article
(This article belongs to the Special Issue Viruses 2024 - A World of Viruses)
Show Figures

Figure 1

Figure 1
<p>Inhibition of apoptosis by CMVs. The MCMV m166 protein inhibits TRAIL expression and HCMV UL141 prevents the expression of TRAIL death receptors on the cell surface. HCMV IE2 interferes with apoptosis signaling by inducing expression of cFLIP, an apoptotic inhibitor. HCMV protein UL36 and its homologous protein M36 in MCMV inhibit extrinsic apoptosis as viral inhibitors of Caspase-8 activation (vICA). HCMV protein UL37x1 and MCMV protein m38.5 are viral mitochondria-localized inhibitors of apoptosis (vMIA), which interfere with BAX. The m41.1 protein of MCMV inhibits apoptosis as a viral inhibitor of BAK oligomerization (vIBO). The HCMV β2.7 non-coding RNA counteracts oxidative stress and maintains the mitochondrial membrane potential. The HCMV protein UL38 inhibits apoptosis by inducing ATF4 accumulation to resolve ER stress. Additionally, UL38 curbs the activation of JNK, which suppresses anti-apoptotic BCL-2 and activates pro-apoptotic BH3 proteins. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Inhibition of necroptosis by CMVs. MCMV protein M45, the viral inhibitor of RIP activation (vIRA), binds and inhibits RIPK3 activation through its RIP homotypic interaction motif (RHIM). M45 thereby inhibits RIPK3-MLKL-mediated necroptosis. HCMV protein UL36 inhibits necroptosis by targeting MLKL and inducing its degradation. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>Inhibition of AIM2 inflammasome-mediated pyroptosis by MCMV. The M84 protein of MCMV interacts with AIM2 and ASC to inhibit inflammasome complex assembly. M84 thereby prevents downstream caspase-1 activation, the release of IL-1β and IL-18, GSDMD cleavage, and pyroptotic cell death. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
Back to TopTop