Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 654
1.
J Virol ; 98(4): e0193523, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38451085

Placental infection plays a central role in the pathogenesis of congenital human cytomegalovirus (HCMV) infections and is a cause of fetal growth restriction and pregnancy loss. HCMV can replicate in some trophoblast cell types, but it remains unclear how the virus evades antiviral immunity in the placenta and how infection compromises placental development and function. Human trophoblast stem cells (TSCs) can be differentiated into extravillous trophoblasts (EVTs), syncytiotrophoblasts (STBs), and organoids, and this study assessed the utility of TSCs as a model of HCMV infection in the first-trimester placenta. HCMV was found to non-productively infect TSCs, EVTs, and STBs. Immunofluorescence assays and flow cytometry experiments further revealed that infected TSCs frequently only express immediate early viral gene products. Similarly, RNA sequencing found that viral gene expression in TSCs does not follow the kinetic patterns observed during lytic infection in fibroblasts. Canonical antiviral responses were largely not observed in HCMV-infected TSCs and TSC-derived trophoblasts. Rather, infection dysregulated factors involved in cell identity, differentiation, and Wingless/Integrated signaling. Thus, while HCMV does not replicate in TSCs, infection may perturb trophoblast differentiation in ways that could interfere with placental function. IMPORTANCE: Placental infection plays a central role in human cytomegalovirus (HCMV) pathogenesis during pregnancy, but the species specificity of HCMV and the limited availability and lifespan of primary trophoblasts have been persistent barriers to understanding how infection impacts this vital organ. Human trophoblast stem cells (TSCs) represent a new approach to modeling viral infection early in placental development. This study reveals that TSCs, like other stem cell types, restrict HCMV replication. However, infection perturbs the expression of genes involved in differentiation and cell fate determination, pointing to a mechanism by which HCMV could cause placental injury.


Cytomegalovirus , Stem Cells , Trophoblasts , Virus Replication , Female , Humans , Pregnancy , Cell Differentiation/genetics , Cell Lineage/genetics , Cytomegalovirus/growth & development , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/virology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Placenta/cytology , Placenta/pathology , Placenta/physiopathology , Placenta/virology , Pregnancy Trimester, First , Stem Cells/cytology , Stem Cells/virology , Trophoblasts/cytology , Trophoblasts/virology
2.
Viruses ; 14(1)2022 01 07.
Article En | MEDLINE | ID: mdl-35062307

Glioblastoma (GBM) is an aggressive primary central nervous system neoplasia with limited therapeutic options and poor prognosis. Following reports of cytomegalovirus (HCMV) in GBM tumors, the anti-viral drug Valganciclovir was administered and found to significantly increase the longevity of GBM patients. While these findings suggest a role for HCMV in GBM, the relationship between them is not clear and remains controversial. Treatment with anti-viral drugs may prove clinically useful; however, their results do not explain the underlying mechanism between HCMV infection and GBM progression. We hypothesized that HCMV infection would metabolically reprogram GBM cells and that these changes would allow for increased tumor progression. We infected LN-18 GBM cells and employed a Seahorse Bioanalyzer to characterize cellular metabolism. Increased mitochondrial respiration and glycolytic rates were observed following infection. These changes were accompanied by elevated production of reactive oxygen species and lactate. Due to lactate's numerous tumor-promoting effects, we examined the impact of paracrine signaling of HCMV-infected GBM cells on uninfected stromal cells. Our results indicated that, independent of viral transmission, the secretome of HCMV-infected GBM cells was able to alter the expression of key metabolic proteins and epigenetic markers. This suggests a mechanism of action where reprogramming of GBM cells alters the surrounding tumor microenvironment to be permissive to tumor progression in a manner akin to the Reverse-Warburg Effect. Overall, this suggests a potential oncomodulatory role for HCMV in the context of GBM.


Cytomegalovirus Infections/physiopathology , Cytomegalovirus/physiology , Glioblastoma/metabolism , Glioblastoma/virology , Paracrine Communication , Secretome , Cell Line, Tumor , Cytomegalovirus/genetics , Cytomegalovirus Infections/virology , Glycolysis , Humans , Lactic Acid/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism , Tumor Microenvironment , Virus Replication
3.
JCI Insight ; 7(1)2022 01 11.
Article En | MEDLINE | ID: mdl-35014624

Congenital cytomegalovirus (cCMV) infection is the leading infectious cause of neurodevelopmental disorders. However, the neuropathogenesis remains largely elusive due to a lack of informative animal models. In this study, we developed a congenital murine CMV (cMCMV) infection mouse model with high survival rate and long survival period that allowed long-term follow-up study of neurodevelopmental disorders. This model involves in utero intracranial injection and mimics many reported clinical manifestations of cCMV infection in infants, including growth restriction, hearing loss, and impaired cognitive and learning-memory abilities. We observed that abnormalities in MRI/CT neuroimaging were consistent with brain hemorrhage and loss of brain parenchyma, which was confirmed by pathological analysis. Neuropathological findings included ventriculomegaly and cortical atrophy associated with impaired proliferation and migration of neural progenitor cells in the developing brain at both embryonic and postnatal stages. Robust inflammatory responses during infection were shown by elevated inflammatory cytokine levels, leukocyte infiltration, and activation of microglia and astrocytes in the brain. Pathological analyses and CT neuroimaging revealed brain calcifications induced by cMCMV infection and cell death via pyroptosis. Furthermore, antiviral treatment with ganciclovir significantly improved neurological functions and mitigated brain damage as shown by CT neuroimaging. These results demonstrate that this model is suitable for investigation of mechanisms of infection-induced brain damage and long-term studies of neurodevelopmental disorders, including the development of interventions to limit CNS damage associated with cCMV infection.


Cytomegalovirus Infections , Disease Models, Animal , Neuroimaging , Animals , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/diagnostic imaging , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/therapy , Female , Follow-Up Studies , Mice , Mice, Inbred ICR , Pregnancy
4.
J Virol ; 96(5): e0182721, 2022 03 09.
Article En | MEDLINE | ID: mdl-35020472

Human cytomegalovirus (HCMV) has a large (∼235 kb) genome with more than 200 predicted open reading frames that exploits numerous cellular factors to facilitate its replication. A key feature of HCMV-infected cells is the emergence of a distinctive membranous cytoplasmic compartment termed the virion assembly compartment (vAC). Here, we report that host protein WD repeat domain 11 (WDR11) plays a key role in vAC formation and virion morphogenesis. We found that WDR11 was upregulated at both mRNA and protein levels during HCMV infection. At the late stage of HCMV replication, WDR11 relocated to the vAC and colocalized with markers of the trans-Golgi network (TGN) and vAC. Depletion of WDR11 hindered HCMV-induced membrane reorganization of the Golgi and TGN, altered vAC formation, and impaired HCMV secondary envelopment and virion morphogenesis. Further, motifs critical for the localization of WDR11 in TGN were identified by alanine-scanning mutagenesis. Mutation of these motifs led to WDR11 mislocation outside the TGN and loss of vAC formation. Taken together, these data indicate that host protein WDR11 is required for efficient viral replication at the stage of virion assembly, possibly by facilitating the remodeling of the endomembrane system for vAC formation and virion morphogenesis. IMPORTANCE During the late phase of human cytomegalovirus (HCMV) infection, the endomembrane system is dramatically reorganized, resulting in the formation of a unique structure termed the virion assembly compartment (vAC), which is critical for the assembly of infectious virions. The mechanism of HCMV-induced vAC formation is still not fully understood. In this report, we identified a host factor, WDR11, that plays an important role in vAC formation. Our findings argue that WDR11 contributes to the relocation of the Golgi and trans-Golgi network to the vAC, a membrane reorganization process that appears to be required for efficient virion maturation. The present work provides new insights into the vAC formation and HCMV virion morphogenesis and a potential novel target for antiviral treatment.


Cytomegalovirus Infections , Cytomegalovirus , Host Microbial Interactions , WD40 Repeats , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/virology , Humans , Morphogenesis , Virion/metabolism , Virus Assembly/genetics , Virus Replication/genetics , WD40 Repeats/genetics , trans-Golgi Network/metabolism
5.
J Virol ; 96(6): e0180621, 2022 03 23.
Article En | MEDLINE | ID: mdl-35045264

Human cytomegalovirus (HCMV) modulates numerous cellular pathways to facilitate infection. Iron is essential to many cellular processes and is often incorporated into proteins and enzymes involved in oxidative phosphorylation and DNA synthesis and repair, among others. Despite its prominent role in the cell, little is known about the regulation of iron metabolism during HCMV infection. Herein, we observe modulation of the transferrin receptor (TfR) during infection and a corresponding change in the cellular labile iron pool. TfR and the iron pool are increased early during infection and then return to mock levels at the late stages of infection. We identified the cellular ubiquitin ligase MARCH1 as an important regulator of TfR. MARCH1 plays a proviral role during infection, as its knockdown leads to a decrease in infectious titers. Knockdown of MARCH1 also leads to an increase in ROS, lipid peroxidation, and mitochondrial dysfunction. Inhibiting an early increase in TfR expression during infection also decreases virus production. These findings indicate the importance of tightly regulating iron metabolism during HCMV infection to facilitate efficient virus production. IMPORTANCE Iron is essential for cells, playing important roles in energy generation, DNA replication, and gene expression. During infection, HCMV alters many cellular processes to aid its replication. We found that iron levels are tightly regulated during infection and that dysregulation of iron levels alters the ability to produce infectious virions. We also found that HCMV inactivates many of the cellular safeguards put in place to deal with excess iron. Thus, infected cells become more susceptible to variations in iron levels, which could be exploited as a therapeutic strategy for dealing with HCMV infections.


Cytomegalovirus Infections , Iron , Ubiquitin-Protein Ligases , Cytomegalovirus/physiology , Cytomegalovirus Infections/enzymology , Cytomegalovirus Infections/physiopathology , Humans , Iron/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
6.
J Hepatol ; 76(1): 115-122, 2022 01.
Article En | MEDLINE | ID: mdl-34563580

BACKGROUND & AIMS: Recent non-malignant non-cirrhotic portal venous system thrombosis (PVT) is a rare condition. Among risk factors for PVT, cytomegalovirus (CMV) disease is usually listed based on a small number of reported cases. The aim of this study was to determine the characteristics and outcomes of PVT associated with CMV disease. METHODS: We conducted a French multicenter retrospective study comparing patients with recent PVT and CMV disease ("CMV positive"; n = 23) to patients with recent PVT for whom CMV testing was negative ("CMV negative"; n = 53) or unavailable ("CMV unknown"; n = 297). RESULTS: Compared to patients from the "CMV negative" and "CMV unknown" groups, patients from the "CMV positive" group were younger, more frequently had fever, and had higher heart rate, lymphocyte count and serum ALT levels (p ≤0.01 for all). The prevalence of immunosuppression did not differ between the 3 groups (4%, 4% and 6%, respectively). Extension of PVT was similar between the 3 groups. Thirteen out of 23 "CMV positive" patients had another risk factor for thrombosis. Besides CMV disease, the number of risk factors for thrombosis was similar between the 3 groups. Heterozygosity for the prothrombin G20210A gene variant was more frequent in "CMV positive" patients (22%) than in the "CMV negative" (4%, p = 0.01) and "CMV unknown" (8%, p = 0.03) groups. Recanalization rate was not influenced by CMV status. CONCLUSIONS: In patients with recent PVT, features of mononucleosis syndrome should raise suspicion of CMV disease. CMV disease does not influence thrombosis extension nor recanalization. More than half of "CMV positive" patients have another risk factor for thrombosis, with a particular link to the prothrombin G20210A gene variant. LAY SUMMARY: Patients with cytomegalovirus (CMV)-associated portal venous system thrombosis have similar thrombosis extension and evolution as patients without CMV disease. However, patients with CMV-associated portal venous system thrombosis more frequently have the prothrombin G20210A gene variant, suggesting that these entities act synergistically to promote thrombosis.


Cytomegalovirus Infections/complications , Portal Vein/abnormalities , Venous Thrombosis/etiology , Adult , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/physiopathology , Female , France , Humans , Male , Middle Aged , Portal Vein/physiopathology , Retrospective Studies , Statistics, Nonparametric , Venous Thrombosis/physiopathology
7.
Placenta ; 117: 200-208, 2022 01.
Article En | MEDLINE | ID: mdl-34933151

INTRODUCTION: Placental dysfunction triggers fetal growth restriction in congenital human cytomegalovirus (HCMV) infection. Studies suggest that HCMV infection interferes with the differentiation of human trophoblasts. However, the underlying mechanisms have not been clarified. This study investigated the impact of HCMV infection on gene transcriptomes in cytotrophoblasts (CTBs) associated with placental dysfunction. METHODS: CTBs were isolated from human term placentas, and spontaneous syncytialization was observed in vitro. The transcriptome profiles were compared between CTB groups with and without HCMV infection by cap analysis gene expression sequencing. The effect of HCMV infection on trophoblast differentiation was evaluated by examining cell fusion status using immunocytochemical staining for desmoplakin and assessing the production of cell differentiation markers, including hCG, PlGF, and soluble Flt-1, using ELISA. RESULTS: The expression of the genes categorized in the signaling pathways related to the cell cycle was significantly enhanced in CTBs with HCMV infection compared with uninfected CTBs. HCMV infection hindered the alteration of the gene expression profile associated with syncytialization. This suppressive effect under HCMV infection was concurrent with the reduction in hCG and PlGF secretion. Immunostaining for desmoplakin revealed that HCMV infection reduced the cell fusion of cultured CTBs. These findings imply that HCMV infection has a negative impact on syncytialization, which is indispensable for the maintenance of villous function. DISCUSSION: HCMV infection interferes with gene expression profiles and functional differentiation of trophoblasts. Suppression of syncytialization may be a survival strategy for HCMV to expand infection and could be associated with placental dysfunction.


Cell Differentiation , Cytomegalovirus Infections/physiopathology , Transcriptome , Trophoblasts/physiology , Cytomegalovirus Infections/metabolism , Female , Humans , Pregnancy , Primary Cell Culture
8.
mBio ; 12(6): e0293421, 2021 12 21.
Article En | MEDLINE | ID: mdl-34903047

Cytomegaloviruses (CMVs) are among the largest pathogenic viruses in mammals. To enable replication of their long double-stranded DNA genomes, CMVs induce profound changes in cell cycle regulation. A hallmark of CMV cell cycle control is the establishment of an unusual cell cycle arrest at the G1/S transition, which is characterized by the coexistence of cell cycle stimulatory and inhibitory activities. While CMVs interfere with cellular DNA synthesis and cell division, they activate S-phase-specific gene expression and nucleotide metabolism. This is facilitated by a set of CMV gene products that target master regulators of G1/S progression such as cyclin E and A kinases, Rb-E2F transcription factors, p53-p21 checkpoint proteins, the APC/C ubiquitin ligase, and the nucleotide hydrolase SAMHD1. While the major themes of cell cycle regulation are well conserved between human and murine CMVs (HCMV and MCMV), there are considerable differences at the level of viral cell cycle effectors and their mechanisms of action. Furthermore, both viruses have evolved unique mechanisms to sense the host cell cycle state and modulate the infection program accordingly. This review provides an overview of conserved and divergent features of G1/S control by MCMV and HCMV.


Cell Cycle Checkpoints , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Muromegalovirus/physiology , Animals , Cytomegalovirus/genetics , G1 Phase , Humans , Mice , Muromegalovirus/genetics , S Phase
9.
mBio ; 12(6): e0226721, 2021 12 21.
Article En | MEDLINE | ID: mdl-34903048

The cGAS/STING/TBK1 (cyclic guanine monophosphate-AMP synthase/stimulator of interferon genes/Tank-binding kinase 1) innate immunity pathway is activated during human cytomegalovirus (HCMV) productive (lytic) replication in fully differentiated cells and during latency within incompletely differentiated myeloid cells. While multiple lytic-phase HCMV proteins neutralize steps along this pathway, none of them are expressed during latency. Here, we show that the latency-associated protein UL138 inhibits the cGAS/STING/TBK1 innate immunity pathway during transfections and infections, in fully differentiated cells and incompletely differentiated myeloid cells, and with loss of function and restoration of function approaches. UL138 inhibits the pathway downstream of STING but upstream of interferon regulatory factor 3 (IRF3) phosphorylation and NF-κB function and reduces the accumulation of interferon beta mRNA during both lytic and latent infections. IMPORTANCE While a cellular restriction versus viral countermeasure arms race between innate immunity and viral latency is expected, few examples have been documented. Our identification of the first HCMV latency protein that inactivates the cGAS/STING/TBK1 innate immune pathway opens the door to understanding how innate immunity, or its neutralization, impacts long-term persistence by HCMV and other latent viruses.


Cytomegalovirus Infections , Cytomegalovirus , Interferon-beta , Membrane Proteins , Virus Latency , Humans , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/virology , Host-Pathogen Interactions , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Latent Infection/genetics , Latent Infection/immunology , Latent Infection/virology , Membrane Proteins/genetics , Membrane Proteins/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
10.
Viruses ; 13(11)2021 11 21.
Article En | MEDLINE | ID: mdl-34835127

Reducing the socioeconomic toll from age-related physical and mental morbidities requires better understanding of factors affecting healthy aging. While many environmental, lifestyle, and genetic factors affect healthy aging, this study addressed the influence of cytomegalovirus (CMV) infection and immunity on age-related inflammation and cognitive abilities. Healthy adults 70-90 years old were recruited into a prospective study investigating relationships between anti-CMV immunity, markers of inflammation, baseline measures of cognitive ability, and changes in cognitive ability over 18 months. Humoral and cellular responses against CMV, levels of inflammatory markers, and cognitive abilities were measured at study entry, with measurement of cognitive abilities repeated 18 months later. CMV-seropositive and -seronegative sub-groups were compared, and relationships between anti-CMV immunity, markers of inflammation, and cognitive ability were assessed. Twenty-eight of 39 participants were CMV-seropositive, and two had CMV-specific CD8+ T cell responses indicative of CMV immune memory inflation. No significant differences for markers of inflammation or measures of cognitive ability were observed between groups, and cognitive scores changed little over 18 months. Significant correlations between markers of inflammation and cognitive scores with interconnection between anti-CMV antibody levels, fractalkine, cognitive ability, and depression scores suggest areas of focus for future studies.


Aging/immunology , Cognition , Cytomegalovirus Infections , Inflammation/immunology , Aged , Aged, 80 and over , Case-Control Studies , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/physiopathology , Female , Humans , Male , Prospective Studies
11.
JCI Insight ; 6(21)2021 11 08.
Article En | MEDLINE | ID: mdl-34609965

CMV causes mostly asymptomatic but lifelong infection. Primary infection or reactivation in immunocompromised individuals can be life-threatening. CMV viremia often occurs in solid organ transplant recipients and associates with decreased graft survival and higher mortality. Furthering understanding of impaired immunity that allows CMV reactivation is critical to guiding antiviral therapy and examining the effect of CMV on solid organ transplant outcomes. This study characterized longitudinal immune responses to CMV in 31 kidney transplant recipients with CMV viremia and matched, nonviremic recipients. Recipients were sampled 3 and 12 months after transplant, with additional samples 1 week and 1 month after viremia. PBMCs were stained for NK and T cell markers. PBMC transcriptomes were characterized by RNA-Seq. Plasma proteins were quantified by Luminex. CD8+ T cell transcriptomes were characterized by single-cell RNA-Seq. Before viremia, patients had high levels of IL-15 with concurrent expansion of immature CD56bright NK cells. After viremia, mature CD56dim NK cells and CD28-CD8+ T cells upregulating inhibitory and NK-associated receptors were expanded. Memory NK cells and NK-like CD28-CD8+ T cells were associated with control of viremia. These findings suggest that signatures of innate activation may be prognostic for CMV reactivation after transplant, while CD8+ T cell functionality is critical for effective control of CMV.


CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/etiology , Kidney Transplantation/adverse effects , Killer Cells, Natural/immunology , Viremia/immunology , Adult , Aged , Cytomegalovirus Infections/physiopathology , Female , Humans , Kidney Transplantation/methods , Male , Middle Aged , Phenotype , Young Adult
12.
Viruses ; 13(8)2021 08 01.
Article En | MEDLINE | ID: mdl-34452384

BACKGROUND: Whether donor (D+) or recipient (R+) cytomegalovirus (CMV) seropositivity is associated with functional impairment in liver transplant recipients is not known. METHODS: Patients included adult liver transplant recipients in the Organ Procurement and Transplantation Network database transplanted over a five-year period from 1 January 2014-31 December 2018. Functional status in the database was assessed using Karnofsky performance scale. A logistic regression model that controlled for potential confounders was used to examine the association of CMV serostatus and functional status. Variables significantly associated with functional status (p < 0.05) were then used to develop propensity score and propensity score matched analysis was conducted where each patient was compared with a matched-control with the same propensity score. RESULTS: Among 30,267 adult liver transplant recipients, D+ or R+ patients had significantly lower functional status at last follow-up than the D-R- cohort (OR 0.88, 95% CI 0.80-0.96, p = 0.007). In propensity score matched model, D+ or R+ patients had significantly lower functional status than matched-controls (p = 0.009). D+ or R+ CMV serostatus (p = 0.018) and low functional level (p < 0.001) were also independently associated with infections as cause-of-death. CONCLUSIONS: D+ or R+ liver transplant recipients had lower functional status and higher risk of deaths due to infections. Future studies are warranted to examine the mechanistic basis of these findings in the setting of transplantation.


Cytomegalovirus Infections/complications , Cytomegalovirus/immunology , Functional Status , Liver Transplantation/adverse effects , Seroepidemiologic Studies , Transplant Recipients/statistics & numerical data , Adult , Aged , Cohort Studies , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/mortality , Cytomegalovirus Infections/physiopathology , Female , Humans , Male , Middle Aged , Propensity Score , Tissue Donors/statistics & numerical data
13.
Medicine (Baltimore) ; 100(31): e26842, 2021 Aug 06.
Article En | MEDLINE | ID: mdl-34397856

INTRODUCTION: Opportunistic infection with multiple pathogens currently has become less uncommon since the application of immunosuppressant or corticosteroid in non- Human immunodeficiency virus patients. However, the clinical diagnosis of the co-infection remains difficult since the uncertainty and deficiency of the microbiologic testing methods. PATIENT CONCERNS: A 66-year-old male patient was admitted to our hospital with chest stuffiness, shortness of breath and elevated body temperature. DIAGNOSIS: He was diagnosed with the co-infection of Pneumocystis jiroveci and cytomegalovirus by metagenomic next-generation sequencing of bronchoalveolar lavage fluid after bronchoscopy. INTERVENTIONS: The patient was empirically treated with broad-spectrum antibiotics, trimethoprim/ sulfamethoxazole and ganciclovir in the beginning of the admission. OUTCOMES: The condition of this patient was not improved even with the intervention at the early stage of the disease. His family requested discharge after 24 inpatient days. LESSONS: This case highlights the application of metagenomic next-generation sequencing in the clinical diagnosis of pulmonary co-infection. Suitable prophylaxis, necessary clinical awareness and accurate diagnosis are indispensable for immunocompromised patients with pulmonary infection.


Bronchoalveolar Lavage Fluid/microbiology , Cytomegalovirus Infections , Cytomegalovirus , Ganciclovir/administration & dosage , Pneumocystis carinii , Pneumonia, Pneumocystis , Trimethoprim, Sulfamethoxazole Drug Combination/administration & dosage , Aged , Anti-Infective Agents/administration & dosage , Bronchoscopy/methods , Coinfection/diagnosis , Coinfection/microbiology , Cytomegalovirus/genetics , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/therapy , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Metagenomics/methods , Methylprednisolone/therapeutic use , Nephrotic Syndrome/drug therapy , Pneumocystis carinii/genetics , Pneumocystis carinii/isolation & purification , Pneumonia, Pneumocystis/diagnosis , Pneumonia, Pneumocystis/physiopathology , Pneumonia, Pneumocystis/therapy
14.
Am J Ophthalmol ; 227: 191-200, 2021 07.
Article En | MEDLINE | ID: mdl-33773985

PURPOSE: To determine distinguishing features of the clinical characteristics of anterior uveitis (AU) caused by herpes simplex virus (HSV), varicella-zoster virus (VZV), and cytomegalovirus (CMV). DESIGN: Retrospective, multicenter case series. METHODS: Consecutive patients with herpetic AU examined at 11 tertiary centers in Japan between January 2012 and December 2017 and who were followed for ≥3 months were evaluated. Diagnosis was made by polymerase chain reaction (PCR) for HSV, VZV, or CMV in the aqueous humor, or classical signs of herpes zoster ophthalmicus. RESULTS: This study enrolled 259 herpetic AU patients, including PCR-proven HSV-AU (30 patients), VZV-AU (50), and CMV-AU (147), and herpes zoster ophthalmicus (32). All HSV-AU and VZV-AU patients were unilateral, while 3% of CMV-AU patients were bilateral. Most HSV-AU and VZV-AU patients were sudden onset with an acute clinical course, while CMV-AU had a more insidious onset and chronic course. There were no significant differences for all surveyed symptoms, signs, and complications between HSV-AU and VZV-AU. However, significant differences were detected for many items between CMV-AU and the other two herpetic AU types. Ocular hyperemia and pain, blurring of vision, ciliary injection, medium-to-large keratic precipitates (KPs), cells and flare in the anterior chamber, and posterior synechia significantly more often occurred in HSV-AU and VZV-AU vs CMV-AU. In contrast, small KPs, coin-shaped KPs, diffuse iris atrophy, elevated intraocular pressure, and glaucoma surgery were significantly more frequent in CMV-AU vs HSV-AU and VZV-AU. CONCLUSION: This multicenter, retrospective study identified distinguishing features of HSV-AU, VZV-AU, and CMV-AU.


Cytomegalovirus Infections/diagnosis , Eye Infections, Viral/diagnosis , Herpes Simplex/diagnosis , Herpes Zoster Ophthalmicus/diagnosis , Uveitis, Anterior/diagnosis , Adult , Aged , Aged, 80 and over , Antiviral Agents/therapeutic use , Aqueous Humor/virology , Cytomegalovirus/genetics , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/virology , Eye Infections, Viral/drug therapy , Eye Infections, Viral/physiopathology , Eye Infections, Viral/virology , Female , Herpes Simplex/drug therapy , Herpes Simplex/physiopathology , Herpes Simplex/virology , Herpes Zoster Ophthalmicus/drug therapy , Herpes Zoster Ophthalmicus/physiopathology , Herpes Zoster Ophthalmicus/virology , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/isolation & purification , Humans , Intraocular Pressure/physiology , Male , Middle Aged , Polymerase Chain Reaction , Retrospective Studies , Simplexvirus/genetics , Simplexvirus/isolation & purification , Uveitis, Anterior/drug therapy , Uveitis, Anterior/physiopathology , Uveitis, Anterior/virology , Visual Acuity/physiology , Young Adult
15.
JAMA Pediatr ; 175(3): e205441, 2021 03 01.
Article En | MEDLINE | ID: mdl-33523119

Importance: The sensitivity of dried blood spots (DBS) to identify newborns with congenital cytomegalovirus (cCMV) infection has not been evaluated in screening studies using the current, higher-sensitivity methods for DBS processing. Objective: To assess the sensitivity of DBS polymerase chain reaction (PCR) for newborn screening for cCMV infection using saliva as the reference standard for screening, followed by collection of a urine sample for confirmation of congenital infection. Design, Setting, and Participants: This population-based cohort study took place at 5 newborn nurseries and 3 neonatal intensive care units in the Minneapolis/Saint Paul area in Minnesota from April 2016 to June 2019. Newborns enrolled with parental consent were screened for cCMV using DBS obtained for routine newborn screening and saliva collected 1 to 2 days after birth. Dried blood spots were tested for CMV DNA by PCR at both the University of Minnesota (UMN) and the US Centers for Disease Control and Prevention (CDC). Saliva swabs were tested by CMV DNA PCR at the UMN laboratory only. Newborns who screened positive by saliva or DBS had a diagnostic urine sample obtained by primary care professionals, tested by PCR within 3 weeks of birth. Analysis began July 2019. Exposures: Detection of CMV from a saliva swab using a PCR assay. Main Outcomes and Measures: Number of children with urine-confirmed cCMV and the proportion of them who were CMV positive through DBS screening. Results: Of 12 554 individuals enrolled through June 2019 (of 25 000 projected enrollment), 56 newborns were confirmed to have cCMV (4.5 per 1000 [95% CI, 3.3-5.7]). Combined DBS results from either UMN or CDC had a sensitivity of 85.7% (48 of 56; 95% CI, 74.3%-92.6%), specificity of 100.0% (95% CI, 100.0%-100.0%), positive predictive value (PPV) of 98.0% (95% CI, 89.3%-99.6%), and negative predictive value (NPV) of 99.9% (95% CI, 99.9%-100.0%). Dried blood spot results from UMN had a sensitivity of 73.2% (95% CI, 60.4%-83.0%), specificity of 100.0% (100.0%-100.0%), PPV of 100.0% (95% CI, 91.4%-100.0%), and NPV of 99.9% (95% CI, 99.8%-99.9%). Dried blood spot results from CDC had a sensitivity of 76.8% (95% CI, 64.2%-85.9%), specificity of 100.0% (95% CI, 100.0%-100.0%), PPV of 97.7% (95% CI, 88.2%-99.6%), and NPV of 99.9% (95% CI, 99.8%-99.9%). Saliva swab results had a sensitivity of 92.9% (52 of 56; 95% CI, 83.0%-97.2%), specificity of 99.9% (95% CI, 99.9%-100.0%), PPV of 86.7% (95% CI, 75.8%-93.1%), and NPV of 100.0% (95% CI, 99.9%-100.0%). Conclusions and Relevance: This study demonstrates relatively high analytical sensitivity for DBS compared with previous studies that performed population-based screening. As more sensitive DNA extraction and PCR methods continue to emerge, DBS-based testing should remain under investigation as a potential low-cost, high-throughput option for cCMV screening.


Cytomegalovirus Infections/diagnosis , Dried Blood Spot Testing/standards , Cohort Studies , Cytomegalovirus Infections/physiopathology , Dried Blood Spot Testing/methods , Dried Blood Spot Testing/statistics & numerical data , Female , Humans , Infant , Infant, Newborn , Male , Minnesota , Neonatal Screening/methods , Prospective Studies , Sensitivity and Specificity
16.
Dig Liver Dis ; 53(6): 722-728, 2021 06.
Article En | MEDLINE | ID: mdl-33441265

BACKGROUND: Cytomegalovirus (CMV) gastritis is occasionally reported in case reports and limited case series. Up to now, it is the largest and most comprehensive retrospective study of CMV gastritis. METHODS: All patients who were histologically diagnosed with CMV gastritis at Linkou Chang Gung Memorial Hospital between January 2000 and April 2020 were included. Patients were divided into two groups according to immunity. Between-group differences in characteristics, manifestations, endoscopic features, prognostic factors, and outcomes were analyzed. The main endpoint was 3-month mortality. RESULTS: A total of 54 patients (34 immunocompromised, 20 immunocompetent) were enrolled. Common presentations included gastrointestinal bleeding (35.2%), abdominal pain (33.3%) and fever (31.5%). The endoscopic features included ulcer (88.9%) and inflammation (11.1%). The 3-month mortality rate was 20.4% and overall mortality rate was 40.7%. Acute kidney injury was the only independent risk factor for 3-month mortality (OR 53.89, 95%CI 1.56-1861.73, p = 0.027). Anti-viral therapy and host immune status did not affect 3-month mortality. CONCLUSION: Both immunocompromised and immunocompetent patients with CMV gastritis have high mortality rates, without significant between-group differences. Acute kidney injury is the only independent predictive factor for 3-month mortality. Prevention of acute kidney injury may possibly improve the 3-month mortality rate.


Cytomegalovirus Infections/mortality , Gastritis/mortality , Adult , Aged , Comorbidity , Cytomegalovirus , Cytomegalovirus Infections/physiopathology , Female , Gastritis/physiopathology , Gastritis/virology , Humans , Immunocompetence , Length of Stay , Male , Middle Aged , Retrospective Studies , Taiwan
17.
Clin Neurol Neurosurg ; 202: 106507, 2021 Mar.
Article En | MEDLINE | ID: mdl-33493883

INTRODUCTION: Polymerase chain reaction (PCR)-based testing of cerebrospinal fluid (CSF) samples has greatly facilitated the diagnosis of central nervous system (CNS) infections. However, the clinical significance of Epstein-Barr virus (EBV) DNA in CSF of individuals with suspected CNS infection remains unclear. We wanted to gain a better understanding of EBV as an infectious agent in immunocompetent patients with CNS disorders. METHODS: We identified cases of EBV-associated CNS infections and reviewed their clinical and laboratory characteristics. The study population was drawn from patients with EBV PCR positivity in CSF who visited Pusan National University Hospital between 2010 and 2019. RESULTS: Of the 780 CSF samples examined during the 10-year study period, 42 (5.4 %) were positive for EBV DNA; 9 of the patients (21.4 %) were diagnosed with non-CNS infectious diseases, such as optic neuritis, Guillain-Barré syndrome, and idiopathic intracranial hypotension, and the other 33 cases were classified as CNS infections (22 as encephalitis and 11 as meningitis). Intensive care unit admission (13/33 patients, 39.3 %) and presence of severe neurological sequelae at discharge (8/33 patients, 24.2 %) were relatively frequent. In 10 patients (30.3 %), the following pathogens were detected in CSF in addition to EBV: varicella-zoster virus (n = 3), cytomegalovirus (n = 2), herpes simplex virus 1 (n = 1), herpes simplex virus 2 (n = 1), Streptococcus pneumomiae (n = 2), and Enterococcus faecalis (n = 1). The EBV-only group (n = 23) and the co-infection group (n = 10) did not differ in age, gender, laboratory data, results of brain imaging studies, clinical manifestations, or prognosis; however, the co-infected patients had higher CSF protein levels. CONCLUSION: EBV DNA in CSF is occasionally found in the immunocompetent population; the virus was commonly associated with encephalitis and poor prognosis, and frequently found together with other microbes in CSF.


DNA, Viral/cerebrospinal fluid , Epstein-Barr Virus Infections/physiopathology , Herpesvirus 4, Human/genetics , Immunocompetence , Infectious Encephalitis/physiopathology , Meningitis/physiopathology , Adult , Aged , Coinfection , Cytomegalovirus Infections/cerebrospinal fluid , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/physiopathology , Encephalitis, Herpes Simplex/cerebrospinal fluid , Encephalitis, Herpes Simplex/complications , Encephalitis, Herpes Simplex/physiopathology , Encephalitis, Viral/cerebrospinal fluid , Encephalitis, Viral/complications , Encephalitis, Viral/physiopathology , Enterococcus faecalis , Epstein-Barr Virus Infections/cerebrospinal fluid , Epstein-Barr Virus Infections/complications , Female , Gram-Positive Bacterial Infections/cerebrospinal fluid , Gram-Positive Bacterial Infections/complications , Gram-Positive Bacterial Infections/physiopathology , Guillain-Barre Syndrome/cerebrospinal fluid , Guillain-Barre Syndrome/complications , Guillain-Barre Syndrome/physiopathology , Humans , Infectious Encephalitis/cerebrospinal fluid , Infectious Encephalitis/complications , Infectious Encephalitis/microbiology , Intensive Care Units , Intracranial Hypotension/cerebrospinal fluid , Intracranial Hypotension/complications , Intracranial Hypotension/physiopathology , Male , Meningitis/cerebrospinal fluid , Meningitis/complications , Meningitis/microbiology , Meningitis, Pneumococcal/cerebrospinal fluid , Meningitis, Pneumococcal/complications , Meningitis, Pneumococcal/physiopathology , Meningitis, Viral/cerebrospinal fluid , Meningitis, Viral/complications , Meningitis, Viral/physiopathology , Middle Aged , Optic Neuritis/cerebrospinal fluid , Optic Neuritis/complications , Optic Neuritis/physiopathology , Streptococcal Infections/cerebrospinal fluid , Streptococcal Infections/complications , Streptococcal Infections/physiopathology , Streptococcus pneumoniae , Varicella Zoster Virus Infection/cerebrospinal fluid , Varicella Zoster Virus Infection/complications
18.
Nat Rev Cardiol ; 18(3): 169-193, 2021 03.
Article En | MEDLINE | ID: mdl-33046850

Inflammatory cardiomyopathy, characterized by inflammatory cell infiltration into the myocardium and a high risk of deteriorating cardiac function, has a heterogeneous aetiology. Inflammatory cardiomyopathy is predominantly mediated by viral infection, but can also be induced by bacterial, protozoal or fungal infections as well as a wide variety of toxic substances and drugs and systemic immune-mediated diseases. Despite extensive research, inflammatory cardiomyopathy complicated by left ventricular dysfunction, heart failure or arrhythmia is associated with a poor prognosis. At present, the reason why some patients recover without residual myocardial injury whereas others develop dilated cardiomyopathy is unclear. The relative roles of the pathogen, host genomics and environmental factors in disease progression and healing are still under discussion, including which viruses are active inducers and which are only bystanders. As a consequence, treatment strategies are not well established. In this Review, we summarize and evaluate the available evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy, with a special focus on virus-induced and virus-associated myocarditis. Furthermore, we identify knowledge gaps, appraise the available experimental models and propose future directions for the field. The current knowledge and open questions regarding the cardiovascular effects associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are also discussed. This Review is the result of scientific cooperation of members of the Heart Failure Association of the ESC, the Heart Failure Society of America and the Japanese Heart Failure Society.


Cardiomyopathies/physiopathology , Inflammation/physiopathology , Myocarditis/physiopathology , Virus Diseases/physiopathology , Animals , Antiviral Agents/therapeutic use , Autoimmunity/immunology , Biopsy , COVID-19/physiopathology , COVID-19/therapy , Cardiomyopathies/diagnosis , Cardiomyopathies/immunology , Cardiomyopathies/therapy , Cardiomyopathy, Dilated , Coronavirus Infections/immunology , Coronavirus Infections/physiopathology , Coronavirus Infections/therapy , Coxsackievirus Infections/immunology , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/therapy , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/therapy , Disease Models, Animal , Echovirus Infections/immunology , Echovirus Infections/physiopathology , Echovirus Infections/therapy , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/physiopathology , Epstein-Barr Virus Infections/therapy , Erythema Infectiosum/immunology , Erythema Infectiosum/physiopathology , Erythema Infectiosum/therapy , HIV Infections/physiopathology , Hepatitis C/immunology , Hepatitis C/physiopathology , Hepatitis C/therapy , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Inflammation/diagnosis , Inflammation/immunology , Inflammation/therapy , Influenza, Human/immunology , Influenza, Human/physiopathology , Influenza, Human/therapy , Leukocytes/immunology , Myocarditis/diagnosis , Myocarditis/immunology , Myocarditis/therapy , Myocardium/pathology , Prognosis , Roseolovirus Infections/immunology , Roseolovirus Infections/physiopathology
19.
J Infect Dis ; 223(4): 655-666, 2021 02 24.
Article En | MEDLINE | ID: mdl-32622351

Cytomegalovirus (CMV) is a major infectious cause of death and disease after transplantation. We have previously demonstrated that the tissue-associated adaptive Vδ2neg γδ T cells are key effectors responding to CMV and associated with recovery, contrasting with their innatelike circulating counterparts, the Vγ9posVδ2pos T cells that respond to phosphoantigens but not to CMV. A third Vγ9negVδ2pos subgroup with adaptive functions has been described in adults. In the current study, we demonstrate that these Vγ9negVδ2pos T cells are also components of the CMV immune response while presenting with distinct characteristics from Vδ2neg γδ T cells. In a cohort of kidney transplant recipients, CMV seropositivity was the unique clinical parameter associated with Vγ9negVδ2pos T-cell expansion and differentiation. Extensive phenotyping demonstrated their substantial cytotoxic potential and activation during acute CMV primary infection or reinfection. In vitro, Vγ9negVδ2pos T cells responded specifically to CMV-infected cells in a T-cell receptor-dependent manner and through strong interferon γ production. Finally, Vγ9negVδ2pos T cells were the only γδ T-cell subset in which expansion was tightly correlated with the severity of CMV disease. To conclude, our results identify a new player in the immune response against CMV and open interesting clinical perspectives for using Vγ9negVδ2pos T cells as an immune marker for CMV disease severity in immunocompromised patients.


Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Biomarkers , Cell Line , Cytomegalovirus Infections/physiopathology , Female , Fibroblasts/immunology , Fibroblasts/virology , Humans , Immunocompromised Host , Interferon-gamma/biosynthesis , Kidney Transplantation , Lymphocyte Activation , Male , Middle Aged , Severity of Illness Index
20.
Augment Altern Commun ; 36(3): 166-178, 2020 09.
Article En | MEDLINE | ID: mdl-33026254

Practitioners need validated strategies for teaching children with significant multiple disabilities (e.g., cognitive, motor, and sensory disability) to use tangible symbols for expressive communication. This single-case experimental design study replicated the positive effect of an adapted protocol for teaching Phase 1 of the Picture Exchange Communication System (PECS) using tangible symbols and extended it to a younger group (4-7 years old) of learners with multiple disabilities. It also tested the effect of an adapted protocol for Phase 2 of PECS to incorporate use of a single switch speech-generating device to gain the attention of an adult communication partner. Two of three students who reached mastery in Phase 1 also reached mastery in Phase 2 and may have generalized requesting behavior from the interventionist (i.e., researchers) to their classroom teacher. Results add to the growing evidence base that shows that the adapted PECS Phase 1 procedures are a promising practice for learners with multiple disabilities, including sensory impairment, and provide preliminary evidence for a more efficient and effective approach to adapting PECS Phase 2 than previously studied for this group of learners. Directions for future research and recommendations for practice are provided.


Communication Aids for Disabled , Communication Disorders/rehabilitation , Developmental Disabilities/rehabilitation , Speech , Child , Child, Preschool , Communication Disorders/physiopathology , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/physiopathology , Developmental Disabilities/physiopathology , Female , Hearing Loss/physiopathology , Humans , Hydrocephalus/physiopathology , Lennox Gastaut Syndrome/physiopathology , Male , Stereotypic Movement Disorder/physiopathology , Vision Disorders/physiopathology
...