Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.973
Filtrar
1.
PLoS Pathog ; 20(9): e1012516, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39236084

RESUMO

ATRX limits the accumulation of human cytomegalovirus (HCMV) Immediate Early (IE) proteins at the start of productive, lytic infections, and thus is a part of the cell-intrinsic defenses against infecting viruses. ATRX is a chromatin remodeler and a component of a histone chaperone complex. Therefore, we hypothesized ATRX would inhibit the transcription of HCMV IE genes by increasing viral genome heterochromatinization and decreasing its accessibility. To test this hypothesis, we quantitated viral transcription and genome structure in cells replete with or depleted of ATRX. We found ATRX did indeed limit viral IE transcription, increase viral genome chromatinization, and decrease viral genome accessibility. The inhibitory effects of ATRX extended to Early (E) and Late (L) viral protein accumulation, viral DNA replication, and progeny virion output. However, we found the negative effects of ATRX on HCMV viral DNA replication were independent of its effects on viral IE and E protein accumulation but correlated with viral genome heterochromatinization. Interestingly, the increased number of viral genomes synthesized in ATRX-depleted cells were not efficiently packaged, indicating the ATRX-mediated restriction to HCMV viral DNA replication may benefit productive infection by increasing viral fitness. Our work mechanistically describes the antiviral function of ATRX and introduces a novel, pro-viral role for this protein, perhaps explaining why, unlike during infections with other herpesviruses, it is not directly targeted by a viral countermeasure in HCMV infected cells.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Replicação do DNA , DNA Viral , Genoma Viral , Heterocromatina , Replicação Viral , Proteína Nuclear Ligada ao X , Humanos , Citomegalovirus/genética , Citomegalovirus/fisiologia , Proteína Nuclear Ligada ao X/genética , Proteína Nuclear Ligada ao X/metabolismo , Heterocromatina/metabolismo , Heterocromatina/genética , DNA Viral/genética , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/genética
2.
Nat Commun ; 15(1): 7640, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223139

RESUMO

Genetic parasites, including viruses and transposons, exploit components from the host for their own replication. However, little is known about virus-transposon interactions within host cells. Here, we discover a strategy where human cytomegalovirus (HCMV) hijacks L1 retrotransposon encoded protein during its replication cycle. HCMV infection upregulates L1 expression by enhancing both the expression of L1-activating transcription factors, YY1 and RUNX3, and the chromatin accessibility of L1 promoter regions. Increased L1 expression, in turn, promotes HCMV replicative fitness. Affinity proteomics reveals UL44, HCMV DNA polymerase subunit, as the most abundant viral binding protein of the L1 ribonucleoprotein (RNP) complex. UL44 directly interacts with L1 ORF2p, inducing DNA damage responses in replicating HCMV compartments. While increased L1-induced mutagenesis is not observed in HCMV for genetic adaptation, the interplay between UL44 and ORF2p accelerates viral DNA replication by alleviating replication stress. Our findings shed light on how HCMV exploits host retrotransposons for enhanced viral fitness.


Assuntos
Citomegalovirus , Replicação do DNA , Elementos Nucleotídeos Longos e Dispersos , Proteínas Virais , Replicação Viral , Humanos , Citomegalovirus/genética , Citomegalovirus/fisiologia , Replicação Viral/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Replicação do DNA/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/genética , Interações Hospedeiro-Patógeno/genética , Retroelementos/genética , Proteínas de Ligação a DNA
3.
J Virol ; 98(9): e0117724, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39194236

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus found in human breast milk that is frequently transmitted from HCMV-seropositive mothers to their infants during the postnatal period. Despite extensive research, the mechanisms underlying HCMV transmission from breast milk and the anatomical location at which virus transfer takes place remain unclear. Breast milk contains many uniquely differentiated macrophages that undergo specific morphological and functional modifications in the mammary gland during lactation. Although the existence of permissive HCMV infection in differentiated macrophages has been well-described, the role of breast milk in this process remains unknown. Herein, we report that exposure of isolated peripheral blood monocytes to breast milk induces their differentiation into macrophages that exhibit an M2 phenotype (CD14highCD163highCD68highCD206high) and promotes a productive and sustained HCMV infection. We also found that breast milk triggers macrophage proliferation and thus sustains a unique population of proliferating, long-lived, and HCMV-susceptible macrophages that are capable of ongoing production of infectious virions. These results suggest a mechanism that explains chronic HCMV shedding into the breast milk of postpartum seropositive mothers. We also found that HCMV virions released from breast milk-induced macrophages generate a productive infection in primary infant tonsil epithelial cells. Collectively, our results suggest that breast milk may facilitate HCMV transmission from mother to infant via the oropharyngeal mucosa. IMPORTANCE: While human cytomegalovirus (HCMV) is frequently detected in the breast milk of HCMV-seropositive women and is often transmitted to infants via breastfeeding, the mechanisms by which this transmission occurs remain unclear. In this study, we modeled HCMV transmission at the oropharyngeal mucosa. We treated human monocytes with breast milk to mimic the lactating mammary gland microenvironment. We found that monocytes differentiated into macrophages with an M2 phenotype, which were highly permissive for HCMV. We also discovered that breast milk induces macrophage proliferation. Thus, exposure to breast milk increased the number of HCMV-susceptible macrophages and supported high levels of infectious HCMV. We found that HCMV virions released from breast milk-induced macrophages could infect primary infant tonsil epithelial cells. Collectively, these findings reveal the dual role of breast milk that induces the differentiation and proliferation of macrophages in the mammary gland and thus facilitates mother-to-child HCMV transmission at the oropharyngeal mucosa.


Assuntos
Diferenciação Celular , Infecções por Citomegalovirus , Citomegalovirus , Macrófagos , Leite Humano , Monócitos , Humanos , Leite Humano/virologia , Macrófagos/virologia , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/transmissão , Citomegalovirus/fisiologia , Feminino , Monócitos/virologia , Células Epiteliais/virologia , Transmissão Vertical de Doenças Infecciosas , Tonsila Palatina/virologia , Tonsila Palatina/citologia , Lactente , Proliferação de Células
4.
J Immunol ; 213(7): 965-970, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39150241

RESUMO

CMV drives the accumulation of virus-specific, highly differentiated CD8 memory T cells (memory inflation [MI]). In mice, MI was shown to directly correlate with the CMV infection dose, yet the CMV-associated CD8 MI plateaus over time. It is unclear how MI is regulated with aging. We infected young mice with 102, 104, and 106 PFU of murine CMV and confirmed that MI magnitude was directly proportional to the infectious dose, reaching a setpoint by midlife. By old age, MI subsided, most prominently in mice infected with 106 PFU, and reached statistical parity between groups in 26-mo-old mice. This corresponded to an age-related loss in lymphatic endothelial cells in lymph nodes, recently shown to be sufficient to drive MI in mice. We propose that MI size and persistence over the lifespan is controlled by the size of the lymphatic endothelial cell niche, whose shrinking leads to reduced MI with aging.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Muromegalovirus , Latência Viral , Animais , Camundongos , Linfócitos T CD8-Positivos/imunologia , Latência Viral/imunologia , Memória Imunológica/imunologia , Muromegalovirus/imunologia , Envelhecimento/imunologia , Camundongos Endogâmicos C57BL , Células T de Memória/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Herpesviridae/imunologia , Células Endoteliais/imunologia , Células Endoteliais/virologia , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Linfonodos/imunologia
5.
Cell Mol Immunol ; 21(9): 959-981, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39134803

RESUMO

Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs. Thus, understanding the pathogenesis of and immune responses to this virus is a prerequisite for developing effective prevention and treatment strategies. Multiple arms of the immune system are engaged to contain the infection, and general concepts of immune control of CMV are now reasonably well understood. Nonetheless, in recent years, tissue-specific immune responses have emerged as an essential factor for resolving CMV infection. As tissues differ in biology and function, so do immune responses to CMV and pathological processes during infection. This review discusses state-of-the-art knowledge of the immune response to CMV infection in tissues, with particular emphasis on several well-studied and most commonly affected organs.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Vigilância Imunológica , Humanos , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Animais , Especificidade de Órgãos/imunologia
6.
Cells ; 13(16)2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39195228

RESUMO

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.


Assuntos
Antivirais , Quinases Ciclina-Dependentes , Citomegalovirus , Proteínas Virais , Humanos , Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Proteínas Virais/metabolismo , Proteínas Virais/química , Quinases Ciclina-Dependentes/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/virologia , Animais , Ciclinas/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)
7.
Nat Commun ; 15(1): 7352, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39187492

RESUMO

The dynamic regulation of mitochondria shape via fission and fusion is critical for cellular responses to stimuli. In homeostatic cells, two modes of mitochondrial fission, midzone and peripheral, provide a decision fork between either proliferation or clearance of mitochondria. However, the relationship between specific mitochondria shapes and functions remains unclear in many biological contexts. While commonly associated with decreased bioenergetics, fragmented mitochondria paradoxically exhibit elevated respiration in several disease states, including infection with the prevalent pathogen human cytomegalovirus (HCMV) and metastatic melanoma. Here, incorporating super-resolution microscopy with mass spectrometry and metabolic assays, we use HCMV infection to establish a molecular mechanism for maintaining respiration within a fragmented mitochondria population. We establish that HCMV induces fragmentation through peripheral mitochondrial fission coupled with suppression of mitochondria fusion. Unlike uninfected cells, the progeny of peripheral fission enter mitochondria-ER encapsulations (MENCs) where they are protected from degradation and bioenergetically stabilized during infection. MENCs also stabilize pro-viral inter-mitochondria contacts (IMCs), which electrochemically link mitochondria and promote respiration. Demonstrating a broader relevance, we show that the fragmented mitochondria within metastatic melanoma cells also form MENCs. Our findings establish a mechanism where mitochondria fragmentation can promote increased respiration, a feature relevant in the context of human diseases.


Assuntos
Citomegalovirus , Retículo Endoplasmático , Metabolismo Energético , Mitocôndrias , Dinâmica Mitocondrial , Humanos , Mitocôndrias/metabolismo , Citomegalovirus/fisiologia , Retículo Endoplasmático/metabolismo , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Melanoma/metabolismo , Melanoma/patologia , Melanoma/virologia , Linhagem Celular Tumoral
8.
Nat Commun ; 15(1): 7353, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39191758

RESUMO

G-quadruplex (G4) structures are found in eukaryotic cell replication origins, but their role in origin function remains unclear. In this study G4 motifs are found in the lytic DNA replication origin (oriLyt) of human cytomegalovirus (HCMV) and recombinant viruses show that a G4 motif in oriLyt essential region I (ER-I) is necessary for viral growth. Replication assays of oriLyt-containing plasmids and biochemical/biophysical analyses show that G4 formation in ER-I is crucial for viral DNA replication. G4 pull-down analysis identifies viral DNA replication factors, such as IE2, UL84, and UL44, as G4-binding proteins. In enzyme-linked immunosorbent assays, specific G4-binding ligands inhibit G4 binding by the viral proteins. The Epstein-Barr virus oriLyt core element also forms a stable G4 that could substitute for the oriLyt ER-I G4 in HCMV. These results demonstrate that viral G4s in replication origins represent an essential structural element in recruiting replication factors and might be a therapeutic target against viral infections.


Assuntos
Citomegalovirus , Replicação do DNA , DNA Viral , Quadruplex G , Proteínas Imediatamente Precoces , Origem de Replicação , Proteínas Virais , Replicação Viral , Citomegalovirus/genética , Citomegalovirus/fisiologia , Humanos , Replicação Viral/genética , Origem de Replicação/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/genética , DNA Viral/genética , DNA Viral/metabolismo , Transativadores/metabolismo , Transativadores/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Ligação Proteica
9.
Viruses ; 16(8)2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39205199

RESUMO

Human cytomegalovirus (HCMV) infection is common in tumor tissues across different types of cancer. While HCMV has not been recognized as a cancer-causing virus, numerous studies hint at its potential role in cancer development where its presence in various cancers corresponds with the hallmarks of cancer. Herein, we discuss and demonstrate that high-risk HCMV-DB and BL strains have the potential to trigger transformation in epithelial cells, including human mammary epithelial cells (HMECs), ovarian epithelial cells (OECs), and prostate epithelial cells (PECs), through the generation of polyploid giant cancer cells (PGCCs). A discussion is provided on how HCMV infection creates a cellular environment that promotes oncogenesis, supporting the continuous growth of CMV-transformed cells. The aforementioned transformed cells, named CTH, CTO, and CTP cells, underwent giant cell cycling with PGCC generation parallel to dedifferentiation, displaying stem-like characteristics and an epithelial-mesenchymal transition (EMT) phenotype. Furthermore, we propose that giant cell cycling through PGCCs, increased EZH2 expression, EMT, and the acquisition of malignant traits represent a deleterious response to the cellular stress induced by high-risk oncogenic HCMV strains, the latter being the origin of the transformation process in epithelial cells upon HCMV infection and leading to adenocarcinoma of poor prognosis.


Assuntos
Transformação Celular Neoplásica , Infecções por Citomegalovirus , Citomegalovirus , Células Epiteliais , Transição Epitelial-Mesenquimal , Células Gigantes , Poliploidia , Humanos , Células Epiteliais/virologia , Citomegalovirus/genética , Citomegalovirus/fisiologia , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/virologia , Células Gigantes/virologia , Feminino , Masculino , Neoplasias/virologia
10.
Viruses ; 16(8)2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39205246

RESUMO

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.


Assuntos
Apoptose , Citomegalovirus , Necroptose , Piroptose , Transdução de Sinais , Replicação Viral , Humanos , Animais , Citomegalovirus/fisiologia , Citomegalovirus/genética , Camundongos , Infecções por Citomegalovirus/virologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune
11.
Viruses ; 16(8)2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39205316

RESUMO

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.


Assuntos
Transdução de Sinais , Viroses , Humanos , Animais , Viroses/metabolismo , Viroses/virologia , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Citomegalovirus/genética
12.
Sci Signal ; 17(851): eadn8727, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39190708

RESUMO

Establishing a nonproductive, quiescent infection within monocytes is essential for the spread of human cytomegalovirus (HCMV). We investigated the mechanisms through which HCMV establishes a quiescent infection in monocytes. US28 is a virally encoded G protein-coupled receptor (GPCR) that is essential for silent infections within cells of the myeloid lineage. We found that preformed US28 was rapidly delivered to monocytes by HCMV viral particles, whereas the de novo synthesis of US28 was delayed for several days. A recombinant mutant virus lacking US28 (US28Δ) was unable to establish a quiescent infection, resulting in a fully productive lytic infection able to produce progeny virus. Infection with US28Δ HCMV resulted in the phosphorylation of the serine and threonine kinase Akt at Ser473 and Thr308, in contrast with the phosphorylation of Akt only at Ser473 after WT viral infection. Inhibiting the dual phosphorylation of Akt prevented the lytic replication of US28Δ, and ectopic expression of a constitutively phosphorylated Akt variant triggered lytic replication of wild-type HCMV. Mechanistically, we found that US28 was necessary and sufficient to attenuate epidermal growth factor receptor (EGFR) signaling induced during the entry of WT virus, which led to the site-specific phosphorylation of Akt at Ser473. Thus, particle-delivered US28 fine-tunes Akt activity by limiting HCMV-induced EGFR activation during viral entry, enabling quiescent infection in monocytes.


Assuntos
Citomegalovirus , Receptores ErbB , Monócitos , Proteínas Proto-Oncogênicas c-akt , Proteínas Virais , Replicação Viral , Citomegalovirus/fisiologia , Citomegalovirus/genética , Citomegalovirus/metabolismo , Humanos , Monócitos/virologia , Monócitos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Fosforilação , Proteínas Virais/metabolismo , Proteínas Virais/genética , Receptores ErbB/metabolismo , Receptores ErbB/genética , Vírion/metabolismo , Vírion/genética , Receptores de Quimiocinas/metabolismo , Receptores de Quimiocinas/genética , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/genética , Transdução de Sinais
13.
Cell Mol Life Sci ; 81(1): 289, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38970696

RESUMO

Congenital human cytomegalovirus (HCMV) infection is a major cause of abnormalities and disorders in the central nervous system (CNS) and/or the peripheral nervous system (PNS). However, the complete pathogenesis of neural differentiation disorders caused by HCMV infection remains to be fully elucidated. Stem cells from human exfoliated deciduous teeth (SHEDs) are mesenchymal stem cells (MSCs) with a high proliferation and neurogenic differentiation capacity. Since SHEDs originate from the neural crest of the early embryonic ectoderm, SHEDs were hypothesized to serve as a promising cell line for investigating the pathogenesis of neural differentiation disorders in the PNS caused by congenital HCMV infection. In this work, SHEDs were demonstrated to be fully permissive to HCMV infection and the virus was able to complete its life cycle in SHEDs. Under neurogenic inductive conditions, HCMV infection of SHEDs caused an abnormal neural morphology. The expression of stem/neural cell markers was also disturbed by HCMV infection. The impairment of neural differentiation was mainly due to a reduction of intracellular cholesterol levels caused by HCMV infection. Sterol regulatory element binding protein-2 (SREBP2) is a critical transcription regulator that guides cholesterol synthesis. HCMV infection was shown to hinder the migration of SREBP2 into nucleus and resulted in perinuclear aggregations of SREBP2 during neural differentiation. Our findings provide new insights into the prevention and treatment of nervous system diseases caused by congenital HCMV infection.


Assuntos
Diferenciação Celular , Colesterol , Infecções por Citomegalovirus , Citomegalovirus , Células-Tronco Mesenquimais , Proteína de Ligação a Elemento Regulador de Esterol 2 , Humanos , Colesterol/metabolismo , Colesterol/biossíntese , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Citomegalovirus/fisiologia , Citomegalovirus/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/virologia , Células-Tronco Mesenquimais/citologia , Células Cultivadas , Dente Decíduo/virologia , Dente Decíduo/citologia , Dente Decíduo/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Neurogênese
14.
Arch Virol ; 169(8): 157, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38969819

RESUMO

Viruses use various strategies and mechanisms to deal with cells and proteins of the immune system that form a barrier against infection. One of these mechanisms is the encoding and production of viral microRNAs (miRNAs), whose function is to regulate the gene expression of the host cell and the virus, thus creating a suitable environment for survival and spreading viral infection. miRNAs are short, single-stranded, non-coding RNA molecules that can regulate the expression of host and viral proteins, and due to their non-immunogenic nature, they are not eliminated by the cells of the immune system. More than half of the viral miRNAs are encoded and produced by Orthoherpesviridae family members. Human cytomegalovirus (HCMV) produces miRNAs that mediate various processes in infected cells to contribute to HCMV pathogenicity, including immune escape, viral latency, and cell apoptosis. Here, we discuss which cellular and viral proteins or cellular pathways and processes these mysterious molecules target to evade immunity and support viral latency in infected cells. We also discuss current evidence that their function of bypassing the host's innate and adaptive immune system is essential for the survival and multiplication of the virus and the spread of HCMV infection.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Evasão da Resposta Imune , MicroRNAs , Latência Viral , Citomegalovirus/genética , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Latência Viral/genética , MicroRNAs/genética , Humanos , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/imunologia , RNA Viral/genética , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética , Regulação Viral da Expressão Gênica
15.
mBio ; 15(8): e0315223, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38953361

RESUMO

There are no licensed vaccines for human cytomegalovirus (HCMV), and current antiviral drugs that target viral proteins are toxic and prone to resistance. Targeting host pathways essential for virus replication provides an alternate strategy that may reduce opportunities for drug resistance to occur. Oxidative stress is triggered by numerous viruses including HCMV. Peroxynitrite is a reactive nitrogen species that is formed during oxidative stress. Herein, we identified that HCMV rapidly induces the generation of intracellular peroxynitrite upon infection in a manner partially dependent upon xanthine oxidase generation. Peroxynitrite promoted HCMV infection in both cell-free and cell-associated infection systems in multiple cell types. Inhibiting peroxynitrite within the first 24 hours of infection prevented HCMV replication and peroxynitrite promoted cell entry and pp65 translocation into the host cell nuclei. Furthermore, using the murine cytomegalovirus model, we demonstrated that antagonizing peroxynitrite significantly reduces cytomegalovirus replication and pathogenesis in vivo. Overall, our study highlights a proviral role for peroxynitrite in CMV infection and implies that RNS and/or the mechanisms that induce their production could be targeted as a novel strategy to inhibit HCMV infection. IMPORTANCE: Human cytomegalovirus (HCMV) causes significant disease in individuals with impaired or immature immune systems, such as transplant patients and after congenital infection. Antiviral drugs that target the virus directly are toxic and are susceptible to antiviral drug resistance due to virus mutations. An alternate strategy is to target processes within host cells that are required by the virus for replication. Herein, we show that HCMV infection triggers a highly reactive molecule, peroxynitrite, during the initial stages of infection. Peroxynitrite was required for the initial entry of the virus into the cell and promotes virus replication in multiple cell types, suggesting a broad pro-viral function. Importantly, targeting peroxynitrite dramatically inhibited cytomegalovirus replication in cells in the laboratory and in mice, suggesting that therapeutic targeting of this molecule and/or the cellular functions it regulates could represent a novel strategy to inhibit HCMV infection.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Modelos Animais de Doenças , Ácido Peroxinitroso , Internalização do Vírus , Replicação Viral , Ácido Peroxinitroso/metabolismo , Ácido Peroxinitroso/farmacologia , Animais , Camundongos , Citomegalovirus/fisiologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/genética , Internalização do Vírus/efeitos dos fármacos , Humanos , Infecções por Citomegalovirus/virologia , Replicação Viral/efeitos dos fármacos , Muromegalovirus/fisiologia , Muromegalovirus/efeitos dos fármacos , Linhagem Celular , Estresse Oxidativo
16.
Antiviral Res ; 230: 105971, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39074588

RESUMO

Human cytomegalovirus (CMV) causes serious developmental disabilities in newborns infected in utero following oral acquisition by the mother. Thus, neutralizing antibodies in maternal saliva have potential to prevent maternal infection and, consequently, fetal transmission and disease. Based on standard cell culture models, CMV entry mediators (and hence neutralizing targets) are cell type-dependent: entry into fibroblasts requires glycoprotein B (gB) and a trimeric complex (TC) of glycoproteins H, L, and O, whereas endothelial and epithelial cell entry additionally requires a pentameric complex (PC) of glycoproteins H and L with UL128, UL130, and UL131A. However, as the mediators of mucosal cell entry and the potential impact of cellular differentiation remained unclear, the present studies utilized mutant viruses, neutralizing antibodies, and soluble TC-receptor to determine the entry mediators required for infection of mucocutaneus cell lines and primary tonsil epithelial cells. Entry into undifferentiated cells was largely PC-dependent, but PC-independent entry could be induced by differentiation. TC-independent entry was also observed and varied by cell line and differentiation. Infection of primary tonsil cells from some donors was entirely TC-independent. In contrast, an antibody to gB or disruption of virion attachment using heparin blocked entry into all cells. These findings indicate that CMV entry into the spectrum of cell types encountered in vivo is likely to be more complex than has been suggested by standard cell culture models and may be influenced by the relative abundance of virion envelope glycoprotein complexes as well as by cell type, tissue of origin, and state of differentiation.


Assuntos
Anticorpos Neutralizantes , Citomegalovirus , Células Epiteliais , Internalização do Vírus , Humanos , Citomegalovirus/fisiologia , Células Epiteliais/virologia , Anticorpos Neutralizantes/imunologia , Linhagem Celular , Proteínas do Envelope Viral/metabolismo , Infecções por Citomegalovirus/virologia , Tonsila Palatina/virologia , Tonsila Palatina/citologia , Células Cultivadas , Diferenciação Celular , Mucosa/virologia , Anticorpos Antivirais/imunologia
17.
Cancer Med ; 13(14): e7402, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39034465

RESUMO

BACKGROUND: Viral reactivations are frequent in hematologial patients due to their cancer-related and drug-induced immunosuppressive status. Daratumumab, an anti-CD38 monoclonal antibody, is used for multiple myeloma (MM) treatment, and causes immunosuppression by targeting CD38-expressing normal lymphocytes. In this single-center two-arm real-life experience, we evaluated incidence of cytomegalovirus (CMV) reactivation in MM patients treated with daratumumab-based regimens as first- or second-line therapy. METHODS: A total of 101 consecutive MM patients were included in this study and were divided into two cohorts: daratumumab and nondaratumumab-based (control) regimens. Patients treated with >2 lines of therapies were excluded to reduce the confounding factor of multi-treated cases. Primary endpoint was the CMV reactivation rate. RESULTS: CMV reactivation rate was significantly higher in the daratumumab cohort compared to control group (33% vs. 4%; p < 0.001), also with higher CMV-DNA levels (>1000 UI/mL in 12% of cases; p < 0.05). However, only one subject developed a CMV disease with severe pneumonia, while 12% of patients were successfully treated with preemptive therapy with valganciclovir. No subjects in the control cohort required anti-CMV agents (p = 0.02). CONCLUSION: Our single-center retrospective experience showed that daratumumab might significantly increase the risk of CMV reactivation in MM, while currently underestimated and related to morbility and mortality in MM patients under treatments. However, further validation on larger and prospective clinical trials are required.


Assuntos
Anticorpos Monoclonais , Infecções por Citomegalovirus , Citomegalovirus , Mieloma Múltiplo , Ativação Viral , Humanos , Mieloma Múltiplo/tratamento farmacológico , Masculino , Feminino , Ativação Viral/efeitos dos fármacos , Idoso , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/efeitos adversos , Citomegalovirus/fisiologia , Pessoa de Meia-Idade , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Estudos Retrospectivos , Valganciclovir/uso terapêutico , Idoso de 80 Anos ou mais , Antivirais/uso terapêutico
18.
Viruses ; 16(7)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-39066247

RESUMO

Despite the significant progress made, CMV infection is one of the most frequent infectious complications in transplant recipients. CMV infections that become refractory or resistant (R/R) to the available antiviral drugs constitute a clinical challenge and are associated with increased morbidity and mortality. Novel anti-CMV therapies have been recently developed and introduced in clinical practice, which may improve the treatment of these infections. In this review, we summarize the treatment options for R/R CMV infections in adult hematopoietic cell transplant and solid organ transplant recipients, with a special focus on newly available antiviral agents with anti-CMV activity, including maribavir and letermovir.


Assuntos
Antivirais , Infecções por Citomegalovirus , Citomegalovirus , Farmacorresistência Viral , Transplantados , Humanos , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/etiologia , Antivirais/uso terapêutico , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Órgãos/efeitos adversos , Acetatos , Diclororribofuranosilbenzimidazol/análogos & derivados , Quinazolinas
19.
Viruses ; 16(7)2024 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-39066333

RESUMO

The complement system is an evolutionarily ancient component of innate immunity that serves as an important first line of defense against pathogens, including viruses. In response to infection, the complement system can be activated by three distinct yet converging pathways (classical, lectin, and alternative) capable of engaging multiple antiviral host responses to confront acute, chronic, and recurrent viral infections. Complement can exert profound antiviral effects via multiple mechanisms including the induction of inflammation and chemotaxis to sites of infection, neutralization/opsonization of viruses and virally infected cells, and it can even shape adaptive immune responses. With millions of years of co-evolution and the ability to establish life-long infections, herpesviruses have evolved unique mechanisms to counter complement-mediated antiviral defenses, thus enabling their survival and replication within humans. This review aims to comprehensively summarize how human herpesviruses engage with the complement system and highlight our understanding of the role of complement in human cytomegalovirus (HCMV) infection, immunity, and viral replication. Herein we describe the novel and unorthodox roles of complement proteins beyond their roles in innate immunity and discuss gaps in knowledge and future directions of complement and HCMV research.


Assuntos
Proteínas do Sistema Complemento , Infecções por Citomegalovirus , Citomegalovirus , Imunidade Inata , Replicação Viral , Humanos , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Proteínas do Sistema Complemento/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Ativação do Complemento/imunologia , Interações Hospedeiro-Patógeno/imunologia , Animais , Imunidade Adaptativa
20.
J Med Virol ; 96(7): e29814, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39015038

RESUMO

Novel biomarkers reflecting the degree of immunosuppression in transplant patients are required to ensure eventual personalized equilibrium between rejection and infection risks. With the above aim, Torque Teno Virus (TTV) viremia was precisely examined in a large cohort of transplanted immunocompromised patients (192 hematological and 60 solid organ transplant recipients) being monitored for Cytomegalovirus reactivation. TTV load was measured in 2612 plasma samples from 448 patients. The results revealed a significant increase in TTV viral load approximately 14 days following CMV reactivation/infection in solid organ transplant (SOT) patients. No recognizable difference in TTV load was noted among hematological patients during the entire timeframe analyzed. Furthermore, a temporal gap of approximately 30 days was noted between the viral load peaks reached by the two viruses, with Cytomegalovirus (CMV) preceding TTV. It was not possible to establish a correlation between CMV reactivation/infection and TTV viremia in hematological patients. On the other hand, the SOT patient cohort allowed us to analyze viral kinetics and draw intriguing conclusions. Taken together, the data suggest, to our knowledge for the first time, that CMV infection itself could potentially cause an increase in TTV load in the peripheral blood of patients undergoing immunosuppressive therapy.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Infecções por Vírus de DNA , Hospedeiro Imunocomprometido , Torque teno virus , Carga Viral , Viremia , Humanos , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/sangue , Masculino , Infecções por Vírus de DNA/virologia , Infecções por Vírus de DNA/sangue , Infecções por Vírus de DNA/imunologia , Pessoa de Meia-Idade , Feminino , Adulto , Terapia de Imunossupressão/efeitos adversos , Ativação Viral , Transplantados/estatística & dados numéricos , Idoso , Estudos de Coortes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA