Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.979
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34115982

RESUMO

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Assuntos
Citomegalovirus/fisiologia , Macrófagos Alveolares/virologia , Animais , Apresentação de Antígeno , Efeito Espectador , Ciclo Celular , Linhagem Celular Transformada , Reprogramação Celular , Citomegalovirus/patogenicidade , Citomegalovirus/ultraestrutura , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Proteínas de Fluorescência Verde/metabolismo , Pulmão/patologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/ultraestrutura , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fenótipo , Células-Tronco/patologia , Replicação Viral/fisiologia , Via de Sinalização Wnt
2.
Cell ; 184(5): 1232-1244.e16, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33626330

RESUMO

Human cytomegalovirus (HCMV) infects the majority of the human population and represents the leading viral cause of congenital birth defects. HCMV utilizes the glycoproteins gHgLgO (Trimer) to bind to platelet-derived growth factor receptor alpha (PDGFRα) and transforming growth factor beta receptor 3 (TGFßR3) to gain entry into multiple cell types. This complex is targeted by potent neutralizing antibodies and represents an important candidate for therapeutics against HCMV. Here, we determine three cryogenic electron microscopy (cryo-EM) structures of the trimer and the details of its interactions with four binding partners: the receptor proteins PDGFRα and TGFßR3 as well as two broadly neutralizing antibodies. Trimer binding to PDGFRα and TGFßR3 is mutually exclusive, suggesting that they function as independent entry receptors. In addition, Trimer-PDGFRα interaction has an inhibitory effect on PDGFRα signaling. Our results provide a framework for understanding HCMV receptor engagement, neutralization, and the development of anti-viral strategies against HCMV.


Assuntos
Citomegalovirus/química , Glicoproteínas de Membrana/química , Proteínas do Envelope Viral/química , Internalização do Vírus , Microscopia Crioeletrônica , Citomegalovirus/fisiologia , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Proteoglicanas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/química , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas do Envelope Viral/metabolismo
3.
Cell ; 174(5): 1158-1171.e19, 2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30057110

RESUMO

Characterizing cell surface receptors mediating viral infection is critical for understanding viral tropism and developing antiviral therapies. Nevertheless, due to challenges associated with detecting protein interactions on the cell surface, the host receptors of many human pathogens remain unknown. Here, we build a library consisting of most single transmembrane human receptors and implement a workflow for unbiased and high-sensitivity detection of receptor-ligand interactions. We apply this technology to elucidate the long-sought receptor of human cytomegalovirus (HCMV), the leading viral cause of congenital birth defects. We identify neuropilin-2 (Nrp2) as the receptor for HCMV-pentamer infection in epithelial/endothelial cells and uncover additional HCMV interactors. Using a combination of biochemistry, cell-based assays, and electron microscopy, we characterize the pentamer-Nrp2 interaction and determine the architecture of the pentamer-Nrp2 complex. This work represents an important approach to the study of host-pathogen interactions and provides a framework for understanding HCMV infection, neutralization, and the development of novel anti-HCMV therapies.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Neuropilina-2/metabolismo , Receptores Virais/metabolismo , Anticorpos Neutralizantes/química , Membrana Celular/metabolismo , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Mapeamento de Epitopos , Feminino , Células HEK293 , Humanos , Conformação Proteica , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus
4.
Nature ; 630(8017): 712-719, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38839957

RESUMO

Genetic screens have transformed our ability to interrogate cellular factor requirements for viral infections1,2, but most current approaches are limited in their sensitivity, biased towards early stages of infection and provide only simplistic phenotypic information that is often based on survival of infected cells2-4. Here, by engineering human cytomegalovirus to express single guide RNA libraries directly from the viral genome, we developed virus-encoded CRISPR-based direct readout screening (VECOS), a sensitive, versatile, viral-centric approach that enables profiling of different stages of viral infection in a pooled format. Using this approach, we identified hundreds of host dependency and restriction factors and quantified their direct effects on viral genome replication, viral particle secretion and infectiousness of secreted particles, providing a multi-dimensional perspective on virus-host interactions. These high-resolution measurements reveal that perturbations altering late stages in the life cycle of human cytomegalovirus (HCMV) mostly regulate viral particle quality rather than quantity, establishing correct virion assembly as a critical stage that is heavily reliant on virus-host interactions. Overall, VECOS facilitates systematic high-resolution dissection of the role of human proteins during the infection cycle, providing a roadmap for in-depth study of host-herpesvirus interactions.


Assuntos
Sistemas CRISPR-Cas , Infecções por Citomegalovirus , Citomegalovirus , Interações Hospedeiro-Patógeno , RNA Guia de Sistemas CRISPR-Cas , Replicação Viral , Humanos , Linhagem Celular , Sistemas CRISPR-Cas/genética , Citomegalovirus/genética , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Genoma Viral/genética , Interações Hospedeiro-Patógeno/genética , RNA Guia de Sistemas CRISPR-Cas/genética , RNA Guia de Sistemas CRISPR-Cas/metabolismo , Vírion/genética , Vírion/metabolismo , Montagem de Vírus/genética , Liberação de Vírus/genética , Replicação Viral/genética
5.
Nature ; 626(8000): 827-835, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38355791

RESUMO

Individuals differ widely in their immune responses, with age, sex and genetic factors having major roles in this inherent variability1-6. However, the variables that drive such differences in cytokine secretion-a crucial component of the host response to immune challenges-remain poorly defined. Here we investigated 136 variables and identified smoking, cytomegalovirus latent infection and body mass index as major contributors to variability in cytokine response, with effects of comparable magnitudes with age, sex and genetics. We find that smoking influences both innate and adaptive immune responses. Notably, its effect on innate responses is quickly lost after smoking cessation and is specifically associated with plasma levels of CEACAM6, whereas its effect on adaptive responses persists long after individuals quit smoking and is associated with epigenetic memory. This is supported by the association of the past smoking effect on cytokine responses with DNA methylation at specific signal trans-activators and regulators of metabolism. Our findings identify three novel variables associated with cytokine secretion variability and reveal roles for smoking in the short- and long-term regulation of immune responses. These results have potential clinical implications for the risk of developing infections, cancers or autoimmune diseases.


Assuntos
Imunidade Adaptativa , Fumar , Feminino , Humanos , Masculino , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/genética , Doenças Autoimunes/etiologia , Doenças Autoimunes/imunologia , Índice de Massa Corporal , Citocinas/sangue , Citocinas/imunologia , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Citomegalovirus/fisiologia , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Infecções/etiologia , Infecções/imunologia , Neoplasias/etiologia , Neoplasias/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fumar/efeitos adversos , Fumar/sangue , Fumar/genética , Fumar/imunologia
6.
Cell ; 157(6): 1460-1472, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24906157

RESUMO

A systematic quantitative analysis of temporal changes in host and viral proteins throughout the course of a productive infection could provide dynamic insights into virus-host interaction. We developed a proteomic technique called "quantitative temporal viromics" (QTV), which employs multiplexed tandem-mass-tag-based mass spectrometry. Human cytomegalovirus (HCMV) is not only an important pathogen but a paradigm of viral immune evasion. QTV detailed how HCMV orchestrates the expression of >8,000 cellular proteins, including 1,200 cell-surface proteins to manipulate signaling pathways and counterintrinsic, innate, and adaptive immune defenses. QTV predicted natural killer and T cell ligands, as well as 29 viral proteins present at the cell surface, potential therapeutic targets. Temporal profiles of >80% of HCMV canonical genes and 14 noncanonical HCMV open reading frames were defined. QTV is a powerful method that can yield important insights into viral infection and is applicable to any virus with a robust in vitro model.


Assuntos
Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Interações Hospedeiro-Patógeno , Proteômica , Virologia/métodos , Humanos , Evasão da Resposta Imune , Células Matadoras Naturais/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Proteínas Virais/análise
7.
Nature ; 621(7977): 120-128, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37558883

RESUMO

Humans display substantial interindividual clinical variability after SARS-CoV-2 infection1-3, the genetic and immunological basis of which has begun to be deciphered4. However, the extent and drivers of population differences in immune responses to SARS-CoV-2 remain unclear. Here we report single-cell RNA-sequencing data for peripheral blood mononuclear cells-from 222 healthy donors of diverse ancestries-that were stimulated with SARS-CoV-2 or influenza A virus. We show that SARS-CoV-2 induces weaker, but more heterogeneous, interferon-stimulated gene activity compared with influenza A virus, and a unique pro-inflammatory signature in myeloid cells. Transcriptional responses to viruses display marked population differences, primarily driven by changes in cell abundance including increased lymphoid differentiation associated with latent cytomegalovirus infection. Expression quantitative trait loci and mediation analyses reveal a broad effect of cell composition on population disparities in immune responses, with genetic variants exerting a strong effect on specific loci. Furthermore, we show that natural selection has increased population differences in immune responses, particularly for variants associated with SARS-CoV-2 response in East Asians, and document the cellular and molecular mechanisms by which Neanderthal introgression has altered immune functions, such as the response of myeloid cells to viruses. Finally, colocalization and transcriptome-wide association analyses reveal an overlap between the genetic basis of immune responses to SARS-CoV-2 and COVID-19 severity, providing insights into the factors contributing to current disparities in COVID-19 risk.


Assuntos
COVID-19 , Genética Populacional , SARS-CoV-2 , Análise da Expressão Gênica de Célula Única , Animais , Humanos , Diferenciação Celular , COVID-19/genética , COVID-19/imunologia , COVID-19/virologia , Citomegalovirus/fisiologia , População do Leste Asiático/genética , Introgressão Genética , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/fisiologia , Interferons/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Células Mieloides/imunologia , Homem de Neandertal/genética , Homem de Neandertal/imunologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Seleção Genética , Latência Viral
8.
Cell ; 151(7): 1569-80, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23260143

RESUMO

Many signaling circuits face a fundamental tradeoff between accelerating their response speed while maintaining final levels below a cytotoxic threshold. Here, we describe a transcriptional circuitry that dynamically converts signaling inputs into faster rates without amplifying final equilibrium levels. Using time-lapse microscopy, we find that transcriptional activators accelerate human cytomegalovirus (CMV) gene expression in single cells without amplifying steady-state expression levels, and this acceleration generates a significant replication advantage. We map the accelerator to a highly self-cooperative transcriptional negative-feedback loop (Hill coefficient ∼7) generated by homomultimerization of the virus's essential transactivator protein IE2 at nuclear PML bodies. Eliminating the IE2-accelerator circuit reduces transcriptional strength through mislocalization of incoming viral genomes away from PML bodies and carries a heavy fitness cost. In general, accelerators may provide a mechanism for signal-transduction circuits to respond quickly to external signals without increasing steady-state levels of potentially cytotoxic molecules.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/genética , Redes Reguladoras de Genes , Aptidão Genética , Citomegalovirus/fisiologia , Retroalimentação Fisiológica , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/metabolismo , Corpos de Inclusão Viral/metabolismo , Imagem com Lapso de Tempo , Transativadores/metabolismo , Ativação Transcricional , Replicação Viral
9.
Proc Natl Acad Sci U S A ; 121(12): e2312290121, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38483999

RESUMO

Human cytomegalovirus (HCMV) infection of monocytes is essential for viral dissemination and persistence. We previously identified that HCMV entry/internalization and subsequent productive infection of this clinically relevant cell type is distinct when compared to other infected cells. We showed that internalization and productive infection required activation of epidermal growth factor receptor (EGFR) and integrin/c-Src, via binding of viral glycoprotein B to EGFR, and the pentamer complex to ß1/ß3 integrins. To understand how virus attachment drives entry, we compared infection of monocytes with viruses containing the pentamer vs. those without the pentamer and then used a phosphoproteomic screen to identify potential phosphorylated proteins that influence HCMV entry and trafficking. The screen revealed that the most prominent pentamer-biased phosphorylated protein was the lipid- and protein-phosphatase phosphatase and tensin homolog (PTEN). PTEN knockdown with siRNA or PTEN inhibition with a PTEN inhibitor decreased pentamer-mediated HCMV entry, without affecting trimer-mediated entry. Inhibition of PTEN activity affected lipid metabolism and interfered with the onset of the endocytic processes required for HCMV entry. PTEN inactivation was sufficient to rescue pentamer-null HCMV from lysosomal degradation. We next examined dephosphorylation of a PTEN substrate Rab7, a regulator of endosomal maturation. Inhibition of PTEN activity prevented dephosphorylation of Rab7. Phosphorylated Rab7, in turn, blocked early endosome to late endosome maturation and promoted nuclear localization of the virus and productive infection.


Assuntos
Monócitos , Internalização do Vírus , Humanos , Células Cultivadas , Monócitos/metabolismo , Citomegalovirus/fisiologia , Receptores ErbB/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo
10.
Proc Natl Acad Sci U S A ; 121(39): e2408078121, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39292744

RESUMO

The Pentamer complex of Human Cytomegalovirus (HCMV) consists of the viral glycoproteins gH, gL, UL128, UL130, and UL131 and is incorporated into infectious virions. HCMV strains propagated extensively in vitro in fibroblasts carry UL128, UL130, or UL131 alleles that do not make a functional complex and thus lack Pentamer function. Adding functional Pentamer to such strains decreases virus growth in fibroblasts. Here, we show that the Pentamer inhibits productive HCMV replication in fibroblasts by repressing viral Immediate Early (IE) transcription. We show that ectopic expression of the viral IE1 protein, a target of Pentamer-mediated transcriptional repression, complements the growth defect of a Pentamer-positive virus. Furthermore, we show that the Pentamer also represses viral IE transcription in cell types where HCMV in vitro latency is studied. Finally, we identify UL130 as a functional subunit of the Pentamer for IE transcriptional repression and demonstrate that cyclic AMP Response Element (CRE) and NFkB sites within the Major Immediate Early Promoter that drives IE1 transcription contribute to this repression. We conclude that the HCMV Pentamer represses viral IE transcription.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Fibroblastos , Proteínas Imediatamente Precoces , Transcrição Gênica , Proteínas do Envelope Viral , Humanos , Citomegalovirus/genética , Citomegalovirus/fisiologia , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/genética , Fibroblastos/virologia , Fibroblastos/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Regulação Viral da Expressão Gênica , Replicação Viral/genética , Glicoproteínas/metabolismo , Glicoproteínas/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Genes Precoces , Regiões Promotoras Genéticas
11.
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
12.
PLoS Pathog ; 20(6): e1012300, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38900818

RESUMO

The AAA-type ATPase VPS4 is recruited by proteins of the endosomal sorting complex required for transport III (ESCRT-III) to catalyse membrane constriction and membrane fission. VPS4A accumulates at the cytoplasmic viral assembly complex (cVAC) of cells infected with human cytomegalovirus (HCMV), the site where nascent virus particles obtain their membrane envelope. Here we show that VPS4A is recruited to the cVAC via interaction with pUL71. Sequence analysis, deep-learning structure prediction, molecular dynamics and mutagenic analysis identify a short peptide motif in the C-terminal region of pUL71 that is necessary and sufficient for the interaction with VPS4A. This motif is predicted to bind the same groove of the N-terminal VPS4A Microtubule-Interacting and Trafficking (MIT) domain as the Type 2 MIT-Interacting Motif (MIM2) of cellular ESCRT-III components, and this viral MIM2-like motif (vMIM2) is conserved across ß-herpesvirus pUL71 homologues. However, recruitment of VPS4A by pUL71 is dispensable for HCMV morphogenesis or replication and the function of the conserved vMIM2 during infection remains enigmatic. VPS4-recruitment via a vMIM2 represents a previously unknown mechanism of molecular mimicry in viruses, extending previous observations that herpesviruses encode proteins with structural and functional homology to cellular ESCRT-III components.


Assuntos
Citomegalovirus , Complexos Endossomais de Distribuição Requeridos para Transporte , Mimetismo Molecular , ATPases Vacuolares Próton-Translocadoras , Montagem de Vírus , Humanos , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , Citomegalovirus/metabolismo , Citomegalovirus/genética , Citomegalovirus/fisiologia , Montagem de Vírus/fisiologia , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética
13.
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
14.
Nature ; 587(7832): 109-114, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32908309

RESUMO

Despite its size and rigidity, the cell nucleus can be moved or reorganized by cytoskeletal filaments under various conditions (for example, during viral infection)1-11. Moreover, whereas chromatin organizes into non-random domains12, extensive heterogeneity at the single-cell level13 means that precisely how and why nuclei reorganize remains an area of intense investigation. Here we describe convolutional neural network-based automated cell classification and analysis pipelines, which revealed the extent to which human cytomegalovirus generates nuclear polarity through a virus-assembled microtubule-organizing centre. Acetylation of tubulin enables microtubules emanating from this centre to rotate the nucleus by engaging cytoplasmically exposed dynein-binding domains in the outer nuclear membrane protein nesprin-2G, which polarizes the inner nuclear membrane protein SUN1. This in turn creates intranuclear polarity in emerin, and thereby controls nuclear actin filaments that spatially segregate viral DNA from inactive histones and host DNA, maximizing virus replication. Our findings demonstrate the extent to which viruses can control the nucleus from the cytoplasm.


Assuntos
Núcleo Celular/metabolismo , Polaridade Celular , Citomegalovirus/fisiologia , Citoplasma/metabolismo , Citoplasma/virologia , Acetilação , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Linhagem Celular , Núcleo Celular/química , DNA Viral/metabolismo , Dineínas/metabolismo , Histonas/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Microtúbulos/química , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Redes Neurais de Computação , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Rotação , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Replicação Viral
15.
Proc Natl Acad Sci U S A ; 120(49): e2309077120, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38011551

RESUMO

Human cytomegalovirus (HCMV) is a paradigm of pathogen immune evasion and sustains lifelong persistent infection in the face of exceptionally powerful host immune responses through the concerted action of multiple immune-evasins. These reduce NK cell activation by inhibiting ligands for activating receptors, expressing ligands for inhibitory receptors, or inhibiting synapse formation. However, these functions only inhibit direct interactions with the infected cell. To determine whether the virus also expresses soluble factors that could modulate NK function at a distance, we systematically screened all 170 HCMV canonical protein-coding genes. This revealed that UL4 encodes a secreted and heavily glycosylated protein (gpUL4) that is expressed with late-phase kinetics and is capable of inhibiting NK cell degranulation. Analyses of gpUL4 binding partners by mass spectrometry identified an interaction with TRAIL. gpUL4 bound TRAIL with picomolar affinity and prevented TRAIL from binding its receptor, thus acting as a TRAIL decoy receptor. TRAIL is found in both soluble and membrane-bound forms, with expression of the membrane-bound form strongly up-regulated on NK cells in response to interferon. gpUL4 inhibited apoptosis induced by soluble TRAIL, while also binding to the NK cell surface in a TRAIL-dependent manner, where it blocked NK cell degranulation and cytokine secretion. gpUL4 therefore acts as an immune-evasin by inhibiting both soluble and membrane-bound TRAIL and is a viral-encoded TRAIL decoy receptor. Interestingly, gpUL4 could also suppress NK responses to heterologous viruses, suggesting that it may act as a systemic virally encoded immunosuppressive agent.


Assuntos
Citomegalovirus , Células Matadoras Naturais , Humanos , Citomegalovirus/fisiologia , Evasão da Resposta Imune , Glicoproteínas/metabolismo , Apoptose
16.
Proc Natl Acad Sci U S A ; 120(33): e2303155120, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37561786

RESUMO

Human cytomegalovirus (HCMV) is a major human pathogen whose life-long persistence is enabled by its remarkable capacity to systematically subvert host immune defenses. In exploring the finding that HCMV infection up-regulates tumor necrosis factor receptor 2 (TNFR2), a ligand for the pro-inflammatory antiviral cytokine TNFα, we found that the underlying mechanism was due to targeting of the protease, A Disintegrin And Metalloproteinase 17 (ADAM17). ADAM17 is the prototype 'sheddase', a family of proteases that cleaves other membrane-bound proteins to release biologically active ectodomains into the supernatant. HCMV impaired ADAM17 surface expression through the action of two virally-encoded proteins in its UL/b' region, UL148 and UL148D. Proteomic plasma membrane profiling of cells infected with an HCMV double-deletion mutant for UL148 and UL148D with restored ADAM17 expression, combined with ADAM17 functional blockade, showed that HCMV stabilized the surface expression of 114 proteins (P < 0.05) in an ADAM17-dependent fashion. These included reported substrates of ADAM17 with established immunological functions such as TNFR2 and jagged1, but also numerous unreported host and viral targets, such as nectin1, UL8, and UL144. Regulation of TNFα-induced cytokine responses and NK inhibition during HCMV infection were dependent on this impairment of ADAM17. We therefore identify a viral immunoregulatory mechanism in which targeting a single sheddase enables broad regulation of multiple critical surface receptors, revealing a paradigm for viral-encoded immunomodulation.


Assuntos
Citomegalovirus , Fator de Necrose Tumoral alfa , Humanos , Citomegalovirus/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Proteoma/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteômica , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Citocinas/metabolismo , Membrana Celular/metabolismo , Metaloproteases/metabolismo , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Virais/metabolismo
17.
J Virol ; 98(4): e0160323, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38526054

RESUMO

mRNA-1647 is an investigational mRNA-based vaccine against cytomegalovirus (CMV) that contains sequences encoding the CMV proteins glycoprotein B and pentamer. Humoral and cellular immune responses were evaluated in blood samples collected from healthy CMV-seropositive and CMV-seronegative adults who participated in a phase 1 trial of a three-dose series of mRNA-1647 (NCT03382405). Neutralizing antibody (nAb) titers against fibroblast and epithelial cell infection in sera from CMV-seronegative mRNA-1647 recipients were higher than those in sera from control CMV-seropositive samples and remained elevated up to 12 months after dose 3. nAb responses elicited by mRNA-1647 were comparable across 14 human CMV (HCMV) strains. Frequencies of antigen-specific memory B cells increased in CMV-seropositive and CMV-seronegative participants after each mRNA-1647 dose and remained elevated for up to 6 months after dose 3. mRNA-1647 elicited robust increases in frequencies and polyfunctionality of CD4+ T helper type 1 and effector CD8+ T cells in samples from CMV-seronegative and CMV-seropositive participants after stimulation with HCMV-specific peptides. The administration of three doses of mRNA-1647 to healthy adults elicited high nAb titers with wide-breadth, long-lasting memory B cells, and strong polyfunctional T-cell responses. These findings support further clinical development of the mRNA-1647 vaccine against CMV.IMPORTANCECytomegalovirus (CMV), a common virus that can infect people of all ages, may lead to serious health problems in unborn babies and those with a weakened immune system. Currently, there is no approved vaccine available to prevent CMV infection; however, the investigational messenger RNA (mRNA)-based CMV vaccine, mRNA-1647, is undergoing evaluation in clinical trials. The current analysis examined samples from a phase 1 trial of mRNA-1647 in healthy adults to better understand how the immune system reacts to vaccination. Three doses of mRNA-1647 produced a long-lasting immune response, thus supporting further investigation of the vaccine in the prevention of CMV infection.CLINICAL TRIALSRegistered at ClinicalTrials.gov (NCT03382405).


Assuntos
Infecções por Citomegalovirus , Vacinas contra Citomegalovirus , Adulto , Humanos , Anticorpos Antivirais , Linfócitos T CD8-Positivos , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/imunologia , Vacinas contra Citomegalovirus/administração & dosagem , Vacinas contra Citomegalovirus/imunologia , RNA Mensageiro/genética
18.
J Virol ; 98(5): e0003224, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38651900

RESUMO

Critical stages of lytic herpes simplex virus type 1 (HSV-1) replication are marked by the sequential expression of immediate early (IE) to early (E), then late (L) viral genes. HSV-1 can also persist in neuronal cells via a non-replicative, transcriptionally repressed infection called latency. The regulation of lytic and latent transcriptional profiles is critical to HSV-1 pathogenesis and persistence. We sought a fluorescence-based approach to observe the outcome of neuronal HSV-1 infection at the single-cell level. To achieve this goal, we constructed and characterized a novel HSV-1 recombinant that enables discrimination between lytic and latent infection. The dual reporter HSV-1 encodes a human cytomegalovirus-immediate early (hCMV-IE) promoter-driven enhanced yellow fluorescent protein (eYFP) to visualize the establishment of infection and an endogenous mCherry-VP26 fusion to report lytic replication. We confirmed that viral gene expression, replication, and spread of infection are not altered by the incorporation of the fluorescent reporters, and fluorescent protein (FP) detection virtuously reports the progression of lytic replication. We demonstrate that the outcome of HSV-1 infection of compartmentalized primary neurons is determined by viral inoculating dose: high-dose axonal inoculation proceeds to lytic replication, whereas low-dose axonal inoculation establishes a latent HSV-1 infection. Interfering with low-dose axonal inoculation via small molecule drugs reports divergent phenotypes of eYFP and mCherry reporter detection, correlating with altered states of viral gene expression. We report that the transcriptional state of neuronal HSV-1 infection is variable in response to changes in the intracellular neuronal environment.IMPORTANCEHerpes simplex virus type 1 (HSV-1) is a prevalent human pathogen that infects approximately 67% of the global human population. HSV-1 invades the peripheral nervous system, where latent HSV-1 infection persists within the host for life. Immunological evasion, viral persistence, and herpetic pathologies are determined by the regulation of HSV-1 gene expression. Studying HSV-1 gene expression during neuronal infection is challenging but essential for the development of antiviral therapeutics and interventions. We used a recombinant HSV-1 to evaluate viral gene expression during infection of primary neurons. Manipulation of cell signaling pathways impacts the establishment and transcriptional state of HSV-1 latency in neurons. The work here provides critical insight into the cellular and viral factors contributing to the establishment of latent HSV-1 infection.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Neurônios , Animais , Humanos , Chlorocebus aethiops , Citomegalovirus/genética , Citomegalovirus/fisiologia , Regulação Viral da Expressão Gênica , Genes Reporter , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Neurônios/virologia , Neurônios/metabolismo , Células Vero , Latência Viral/genética , Replicação Viral
19.
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
20.
J Virol ; 98(3): e0127823, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38345384

RESUMO

Cytomegalovirus (CMV), a type of herpes virus, is the predominant cause of congenital anomalies due to intrauterine infections in humans. Adverse outcomes related to intrauterine infections with human cytomegalovirus (HCMV) vary widely, depending on factors such as fetal infection timing, infection route, and viral virulence. The precise mechanism underlying HCMV susceptibility remains unclear. In this study, we compared the susceptibility of neonatal human dermal fibroblast cells (NHDFCs) and human induced pluripotent stem cells (hiPSCs) derived from NHDFCs, which are genetically identical to HCMV, using immunostaining, microarray, in situ hybridization, quantitative PCR, and scanning electron microscopy. These cells were previously used to compare CMV susceptibility, but the underlying mechanisms were not fully elucidated. HCMV susceptibility of hiPSCs was significantly lower in the earliest phase. No shared gene ontologies were observed immediately post-infection between the two cell types using microarray analysis. Early-stage expression of HCMV antigens and the HCMV genome was minimal in immunostaining and in in situ hybridization in hiPSCs. This strongly suggests that HCMV does not readily bind to hiPSC surfaces. Scanning electron microscopy performed using the NanoSuit method confirmed the scarcity of HCMV particles on hiPSC surfaces. The zeta potential and charge mapping of the charged surface in NHDFCs and hiPSCs exhibited minimal differences when assessed using zeta potential analyzer and scanning ion conductance microscopy; however, the expression of heparan sulfate (HS) was significantly lower in hiPSCs compared with that in NHDFCs. Thus, HS expression could be a primary determinant of HCMV resistance in hiPSCs at the attachment level. IMPORTANCE: Numerous factors such as attachment, virus particle entry, transcription, and virus particle egress can affect viral susceptibility. Since 1984, pluripotent cells are known to be CMV resistant; however, the exact mechanism underlying this resistance remains elusive. Some researchers suggest inhibition in the initial phase of HCMV binding, while others have suggested the possibility of a sufficient amount of HCMV entering the cells to establish latency. This study demonstrates that HCMV particles rarely attach to the surfaces of hiPSCs. This is not due to limitations in the electrostatic interactions between the surface of hiPSCs and HCMV particles, but due to HS expression. Therefore, HS expression should be recognized as a key factor in determining the susceptibility of HCMV in congenital infection in vitro and in vivo. In the future, drugs targeting HS may become crucial for the treatment of congenital CMV infections. Thus, further research in this area is warranted.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Heparitina Sulfato , Células-Tronco Pluripotentes Induzidas , Humanos , Recém-Nascido , Membrana Celular/química , Membrana Celular/metabolismo , Citomegalovirus/fisiologia , Heparitina Sulfato/análise , Heparitina Sulfato/metabolismo , Infecções por Herpesviridae , Células-Tronco Pluripotentes Induzidas/química , Células-Tronco Pluripotentes Induzidas/metabolismo , Fibroblastos/química , Fibroblastos/metabolismo , Fibroblastos/virologia , Pele/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA