Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Gastroenterology ; 159(1): 214-226.e1, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32247021

RESUMO

BACKGROUND & AIMS: Intestinal microfold (M) cells are a unique subset of intestinal epithelial cells in the Peyer's patches that regulate mucosal immunity, serving as portals for sampling and uptake of luminal antigens. The inability to efficiently develop human M cells in cell culture has impeded studies of the intestinal immune system. We aimed to identify signaling pathways required for differentiation of human M cells and establish a robust culture system using human ileum enteroids. METHODS: We analyzed transcriptome data from mouse Peyer's patches to identify cell populations in close proximity to M cells. We used the human enteroid system to determine which cytokines were required to induce M-cell differentiation. We performed transcriptome, immunofluorescence, scanning electron microscope, and transcytosis experiments to validate the development of phenotypic and functional human M cells. RESULTS: A combination of retinoic acid and lymphotoxin induced differentiation of glycoprotein 2-positive human M cells, which lack apical microvilli structure. Upregulated expression of innate immune-related genes within M cells correlated with a lack of viral antigens after rotavirus infection. Human M cells, developed in the enteroid system, internalized and transported enteric viruses, such as rotavirus and reovirus, across the intestinal epithelium barrier in the enteroids. CONCLUSIONS: We identified signaling pathways required for differentiation of intestinal M cells, and used this information to create a robust culture method to develop human M cells with capacity for internalization and transport of viruses. Studies of this model might increase our understanding of antigen presentation and the systemic entry of enteric pathogens in the human intestine.


Assuntos
Diferenciação Celular/imunologia , Linfotoxina-alfa/metabolismo , Nódulos Linfáticos Agregados/imunologia , Transdução de Sinais/imunologia , Tretinoína/metabolismo , Animais , Apresentação de Antígeno/imunologia , Técnicas de Cultura de Células/métodos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Humanos , Íleo/citologia , Íleo/imunologia , Imunidade nas Mucosas , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Camundongos , NF-kappa B/metabolismo , Organoides , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/metabolismo , Cultura Primária de Células , Proteínas Recombinantes/metabolismo
2.
J Virol ; 92(4)2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29167347

RESUMO

The viral Bcl-2 homolog (vBcl2) of Kaposi's sarcoma-associated herpesvirus (KSHV) displays efficient antiapoptotic and antiautophagic activity through its central BH3 domain, which functions to prolong the life span of virus-infected cells and ultimately enhances virus replication and latency. Independent of its antiapoptotic and antiautophagic activity, vBcl2 also plays an essential role in KSHV lytic replication through its amino-terminal amino acids (aa) 11 to 20. Here, we report a novel molecular mechanism of vBcl2-mediated regulation of KSHV lytic replication. vBcl2 specifically bound the tegument protein open reading frame 55 (ORF55) through its amino-terminal aa 11 to 20, allowing their association with virions. Consequently, the vBcl2 peptide derived from vBcl2 aa 11 to 20 effectively disrupted the interaction between vBcl2 and ORF55, inhibiting the incorporation of the ORF55 tegument protein into virions. This study provides new insight into vBcl2's function in KSHV virion assembly that is separable from its inhibitory role in host apoptosis and autophagy.IMPORTANCE KSHV, an important human pathogen accounting for a large percentage of virally caused cancers worldwide, has evolved a variety of stratagems for evading host immune responses to establish lifelong persistent infection. Upon viral infection, infected cells can go through programmed cell death, including apoptosis and autophagy, which plays an effective role in antiviral responses. To counter the host response, KSHV vBcl2 efficiently blocks apoptosis and autophagy to persist for the life span of virus-infected cells. Besides its anti-programmed-cell-death activity, vBcl2 also interacts with the ORF55 tegument protein for virion assembly in infected cells. Interestingly, the vBcl2 peptide disrupts the vBcl2-ORF55 interaction and effectively inhibits KSHV virion assembly. This study indicates that KSHV vBcl2 harbors at least three genetically separable functions to modulate both host cell death signaling and virion production and that the vBcl2 peptide can be developed as an anti-KSHV therapeutic application.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteínas Oncogênicas/fisiologia , Fases de Leitura Aberta , Proteínas Virais/fisiologia , Montagem de Vírus , Apoptose , Autofagia , Sequência de Bases , Replicação do DNA , DNA Viral/genética , Expressão Gênica , Técnicas de Inativação de Genes , Genoma Viral , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Proteínas Oncogênicas/genética , Proteínas Virais/genética
3.
J Virol ; 90(17): 7657-66, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27307571

RESUMO

UNLABELLED: The K1 gene product of Kaposi's sarcoma-associated herpesvirus (KSHV) is encoded by the first open reading frame (ORF) of the viral genome. To investigate the role of the K1 gene during the KSHV life cycle, we constructed a set of recombinant viruses that contained either wild-type (WT) K1, a deleted K1 ORF (KSHVΔK1), stop codons within the K1 ORF (KSHV-K15×STOP), or a revertant K1 virus (KSHV-K1REV). We report that the recombinant viruses KSHVΔK1 and KSHV-K15×STOP displayed significantly reduced lytic replication compared to WT KSHV and KSHV-K1REV upon reactivation from latency. Additionally, cells infected with the recombinant viruses KSHVΔK1 and KSHV-K15×STOP also yielded smaller amounts of infectious progeny upon reactivation than did WT KSHV- and KSHV-K1REV-infected cells. Upon reactivation from latency, WT KSHV- and KSHV-K1REV-infected cells displayed activated Akt kinase, as evidenced by its phosphorylation, while cells infected with viruses deleted for K1 showed reduced phosphorylation and activation of Akt kinase. Overall, our results suggest that K1 plays an important role during the KSHV life cycle. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of three human malignancies, and KSHV K1 is a signaling protein that has been shown to be involved in cellular transformation and to activate the phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR pathway. In order to investigate the role of the K1 protein in the life cycle of KSHV, we constructed recombinant viruses that were deficient for K1. We found that K1 deletion viruses displayed reduced lytic replication compared to the WT virus and also yielded smaller numbers of infectious progeny. We report that K1 plays an important role in the life cycle of KSHV.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteínas Virais/metabolismo , Replicação Viral , Linhagem Celular , Códon sem Sentido , Deleção de Genes , Herpesvirus Humano 8/genética , Humanos , Supressão Genética , Proteínas Virais/genética
4.
J Virol ; 90(8): 3849-3859, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26819313

RESUMO

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) has tropism for B lymphocytes, in which it establishes latency, and can also cause lymphoproliferative disorders of these cells manifesting as primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). T cell immunity is vital for the control of KSHV infection and disease; however, few models of B lymphocyte infection exist to study immune recognition of such cells. Here, we developed a model of B lymphocyte infection with KSHV in which infected tonsillar B lymphocytes were expanded by providing mitogenic stimuli and then challenged with KSHV-specific CD4(+)T cells. The infected cells expressed viral proteins found in PELs, namely, LANA and viral IRF3 (vIRF3), albeit at lower levels, with similar patterns of gene expression for the major latency, viral interleukin 6 (vIL-6), and vIRF3 transcripts. Despite low-level expression of open reading frame 50 (ORF50), transcripts for the immune evasion genes K3 and K5 were detected, with some downregulation of cell surface-expressed CD86 and ICAM. The vast majority of infected lymphocytes expressed IgM heavy chains with Igλ light chains, recapitulating the features seen in infected cells in MCD. We assessed the ability of the infected lymphocytes to be targeted by a panel of major histocompatibility complex (MHC) class II-matched CD4(+)T cells and found that LANA-specific T cells restricted to different epitopes recognized these infected cells. Given that at least some KSHV latent antigens are thought to be poor targets for CD8(+)T cells, we suggest that CD4(+)T cells are potentially important effectors for thein vivocontrol of KSHV-infected B lymphocytes. IMPORTANCE: KSHV establishes a latent reservoir within B lymphocytes, but few models exist to study KSHV-infected B cells other than the transformed PEL cell lines, which have likely accrued mutations during the transformation process. We developed a model of KSHV-infected primary B lymphocytes that recapitulates features seen in PEL and MCD by gene expression and cell phenotype analysis, allowing the study of T cell recognition of these cells. Challenge of KSHV-infected B cells with CD4(+)T cells specific for LANA, a protein expressed in all KSHV-infected cells and malignanciesin vivo, showed that these effectors could efficiently recognize such targets. Given that the virus expresses immune evasion genes or uses proteins with intrinsic properties, such as LANA, that minimize epitope recognition by CD8(+)T cells, CD4(+)T cell immunity to KSHV may be important for maintaining the virus-host balance.


Assuntos
Antígenos Virais/imunologia , Linfócitos B/virologia , Linfócitos T CD4-Positivos/imunologia , Transformação Celular Viral , Herpesvirus Humano 8/fisiologia , Proteínas Nucleares/imunologia , Antígenos de Superfície/imunologia , Proliferação de Células , Células Cultivadas , Expressão Gênica , Genes Virais , Herpesvirus Humano 8/genética , Humanos , Fatores Reguladores de Interferon/imunologia , Modelos Biológicos , Tonsila Palatina/citologia , Fenótipo , Receptores Imunológicos/biossíntese , Proteínas Virais/imunologia
5.
J Virol ; 89(10): 5298-307, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740992

RESUMO

UNLABELLED: The Kaposi's sarcoma-associated herpesvirus (KSHV) open reading frame 16 (orf16) encodes a viral Bcl-2 (vBcl-2) protein which shares sequence and functional homology with the Bcl-2 family. Like its cellular homologs, vBcl-2 protects various cell types from apoptosis and can also negatively regulate autophagy. vBcl-2 is transcribed during lytic infection; however, its exact function has not been determined to date. By using bacterial artificial chromosome 16 (BAC16) clone carrying the full-length KSHV genome, we have generated recombinant KSHV mutants that fail to express vBcl-2 or express mCherry-tagged vBcl-2. We show that the vBcl-2 protein is expressed at relatively low levels during lytic induction and that a lack of vBcl-2 largely reduces the efficiency of KSHV reactivation in terms of lytic gene expression, viral DNA replication, and production of infectious particles. In contrast, the establishment of latency was not affected by the absence of vBcl-2. Our findings suggest an important role for vBcl-2 during initial phases of lytic reactivation and/or during subsequent viral propagation. Given the known functions of vBcl-2 in regulating apoptosis and autophagy, which involve its direct interaction with cellular proteins and thus require high levels of protein expression, it appears that vBcl-2 may have additional regulatory functions that do not depend on high levels of protein expression. IMPORTANCE: The present study shows for the first time the expression of endogenous vBcl-2 protein in KSHV-infected cell lines and demonstrates the importance of vBcl-2 during the initial phases of lytic reactivation and/or during its subsequent propagation. It is suggested that vBcl-2 has additional regulatory functions beyond apoptosis and autophagy repression that do not depend on high levels of protein expression.


Assuntos
Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/fisiologia , Proteínas Virais/genética , Proteínas Virais/fisiologia , Ativação Viral/genética , Ativação Viral/fisiologia , Sequência de Bases , Linhagem Celular , Cromossomos Artificiais Bacterianos/genética , DNA Recombinante/genética , DNA Viral/genética , Expressão Gênica , Genes Virais , Células HEK293 , Herpesvirus Humano 8/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Dados de Sequência Molecular , Mutação , Recombinação Genética , Replicação Viral
6.
J Virol ; 89(10): 5308-17, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740994

RESUMO

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) evades host defenses through tight suppression of autophagy by targeting each step of its signal transduction: by viral Bcl-2 (vBcl-2) in vesicle nucleation, by viral FLIP (vFLIP) in vesicle elongation, and by K7 in vesicle maturation. By exploring the roles of KSHV autophagy-modulating genes, we found, surprisingly, that vBcl-2 is essential for KSHV lytic replication, whereas vFLIP and K7 are dispensable. Knocking out vBcl-2 from the KSHV genome resulted in decreased lytic gene expression at the mRNA and protein levels, a lower viral DNA copy number, and, consequently, a dramatic reduction in the amount of progeny infectious viruses, as also described in the accompanying article (A. Gelgor, I. Kalt, S. Bergson, K. F. Brulois, J. U. Jung, and R. Sarid, J Virol 89:5298-5307, 2015). More importantly, the antiapoptotic and antiautophagic functions of vBcl-2 were not required for KSHV lytic replication. Using a comprehensive mutagenesis analysis, we identified that glutamic acid 14 (E14) of vBcl-2 is critical for KSHV lytic replication. Mutating E14 to alanine totally blocked KSHV lytic replication but showed little or no effect on the antiapoptotic and antiautophagic functions of vBcl-2. Our study indicates that vBcl-2 harbors at least three important and genetically separable functions to modulate both cellular signaling and the virus life cycle. IMPORTANCE: The present study shows for the first time that vBcl-2 is essential for KSHV lytic replication. Removal of the vBcl-2 gene results in a lower level of KSHV lytic gene expression, impaired viral DNA replication, and consequently, a dramatic reduction in the level of progeny production. More importantly, the role of vBcl-2 in KSHV lytic replication is genetically separated from its antiapoptotic and antiautophagic functions, suggesting that the KSHV Bcl-2 carries a novel function in viral lytic replication.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteínas Oncogênicas/fisiologia , Proteínas Virais/fisiologia , Replicação Viral/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Apoptose , Autofagia , Sequência de Bases , Linhagem Celular , Replicação do DNA , DNA Viral/genética , Expressão Gênica , Técnicas de Inativação de Genes , Genoma Viral , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/fisiologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Oncogênicas/antagonistas & inibidores , Proteínas Oncogênicas/genética , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética , Replicação Viral/genética
7.
J Virol ; 88(11): 6512-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24623444

RESUMO

The Kaposi's sarcoma-associated herpesvirus (KSHV) ORF36 protein kinase is translated as a downstream gene from the ORF35-37 polycistronic mRNA via a unique mechanism involving short upstream open reading frames (uORFs) located in the 5' untranslated region. Here, we confirm that ORF35-37 is functionally dicistronic during infection and demonstrate that mutation of the dominant uORF restricts KSHV replication. Leaky scanning past the uORFs facilitates ORF35 expression, while a reinitiation mechanism after translation of the uORFs enables ORF36 expression.


Assuntos
Regulação Viral da Expressão Gênica/genética , Herpesvirus Humano 8/genética , Fases de Leitura Aberta/genética , Proteínas Quinases/metabolismo , RNA Mensageiro/genética , Herpesvirus Humano 8/metabolismo , Modelos Genéticos , Proteínas Quinases/genética , RNA Mensageiro/metabolismo
8.
J Virol ; 88(21): 12839-52, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25165104

RESUMO

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is a cancer-related human virus, classified as a member of the Gammaherpesvirinae subfamily. We report here the construction of a dual fluorescent-tagged KSHV genome (BAC16-mCherry-ORF45), which constitutively expresses green fluorescent protein (GFP) and contains the tegument multifunctional ORF45 protein as a fusion protein with monomeric Cherry fluorescent protein (mCherry). We confirmed that this virus is properly expressed and correctly replicates and that the mCherry-ORF45 protein is incorporated into the virions. Using this labeled virus, we describe the dynamics of mCherry-ORF45 expression and localization in newly infected cells as well as in latently infected cells undergoing lytic induction and show that mCherry can be used to monitor cells undergoing the lytic viral cycle. This virus is likely to enable future studies monitoring the dynamics of viral trafficking and tegumentation during viral ingress and egress. IMPORTANCE: The present study describes the construction and characterization of a new recombinant KSHV genome BAC16 clone which expresses mCherry-tagged ORF45. This virus enables the tracking of cells undergoing lytic infection and can be used to address issues related to the trafficking and maturation pathways of KSHV virions.


Assuntos
Citosol/química , Citosol/virologia , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/análise , Fusão Gênica Artificial , Linhagem Celular , Perfilação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Coloração e Rotulagem/métodos , Latência Viral , Replicação Viral
9.
J Virol ; 88(19): 11369-82, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25056896

RESUMO

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with several human malignances. As saliva is likely the major vehicle for KSHV transmission, we studied in vitro KSHV infection of oral epithelial cells. Through infection of two types of oral epithelial cells, normal human oral keratinocytes (NHOKs) and papilloma-immortalized human oral keratinocyte (HOK16B) cells, we found that KSHV can undergo robust lytic replication in oral epithelial cells. By employing de novo lytic infection of HOK16B cells, we studied the functions of two previously uncharacterized genes, ORF18 and ORF30, during the KSHV lytic cycle. For this purpose, an ORF18-deficient virus and an ORF30-deficient virus were generated using a mutagenesis strategy based on bacterial artificial chromosome (BAC) technology. We found that neither ORF18 nor ORF30 is required for immediately early or early gene expression or viral DNA replication, but each is essential for late gene expression during both de novo lytic replication and reactivation. This critical role of ORF18 and ORF30 in late gene expression was also observed during KSHV reactivation. In addition, global analysis of viral transcripts by RNA sequencing indicated that ORF18 and ORF30 control the same set of viral genes. Therefore, we suggest that these two viral ORFs are involved in the same mechanism or pathway that coregulates the viral late genes as a group. IMPORTANCE: While KSHV can infect multiple cell types in vitro, only a few can support a full lytic replication cycle with progeny virions produced. Consequently, KSHV lytic replication is mostly studied through reactivation, which requires chemicals to induce the lytic cycle or overexpression of the viral transcriptional activator, RTA. In this study, we present a robust de novo lytic infection system based on oral epithelial cells. Using this system, we demonstrate the role of two viral ORFs, ORF18 and ORF30, in regulating viral gene expression during KSHV lytic replication. As the major route of KSHV transmission is thought to be via saliva, this new KSHV lytic replication system will have important utility in the field.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Queratinócitos/virologia , Fases de Leitura Aberta , Proteínas Virais/genética , Sequência de Bases , Linhagem Celular , Cromossomos Artificiais Bacterianos , Herpesvirus Humano 8/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Queratinócitos/patologia , Dados de Sequência Molecular , Mucosa Bucal/patologia , Mucosa Bucal/virologia , Deleção de Sequência , Transdução de Sinais , Proteínas Virais/metabolismo , Ativação Viral , Replicação Viral
10.
PLoS Pathog ; 9(1): e1003156, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23382684

RESUMO

The Kaposi's sarcoma-associated herpesvirus (KSHV) protein kinase, encoded by ORF36, functions to phosphorylate cellular and viral targets important in the KSHV lifecycle and to activate the anti-viral prodrug ganciclovir. Unlike the vast majority of mapped KSHV genes, no viral transcript has been identified with ORF36 positioned as the 5'-proximal gene. Here we report that ORF36 is robustly translated as a downstream cistron from the ORF35-37 polycistronic transcript in a cap-dependent manner. We identified two short, upstream open reading frames (uORFs) within the 5' UTR of the polycistronic mRNA. While both uORFs function as negative regulators of ORF35, unexpectedly, the second allows for the translation of the downstream ORF36 gene by a termination-reinitiation mechanism. Positional conservation of uORFs within a number of related viruses suggests that this may be a common γ-herpesviral adaptation of a host translational regulatory mechanism.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Fases de Leitura Aberta/genética , RNA Mensageiro/genética , RNA Viral/genética , Regiões 5' não Traduzidas/genética , Sequência de Bases , Linhagem Celular , Humanos , Dados de Sequência Molecular , Família Multigênica , Biossíntese de Proteínas , RNA Viral/química , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica
11.
J Virol ; 87(22): 12499-503, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24027317

RESUMO

Autophagy is an important innate safeguard mechanism for protecting an organism against invasion by pathogens. We have previously discovered that Kaposi's sarcoma-associated herpesvirus (KSHV) evades this host defense through tight suppression of autophagy by targeting multiple steps of autophagy signal transduction. Here, we report that KSHV K7 protein interacts with Rubicon autophagy protein and inhibits the autophagosome maturation step by blocking Vps34 enzymatic activity, further highlighting how KSHV deregulates autophagy-mediated host immunity for its life cycle.


Assuntos
Autofagia , Herpesvirus Humano 8/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Mitocondriais/metabolismo , Fagossomos/metabolismo , Sarcoma de Kaposi/patologia , Proteínas Virais/metabolismo , Proteínas Relacionadas à Autofagia , Células HeLa , Humanos , Immunoblotting , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais
12.
Res Sq ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38947054

RESUMO

Stromal cells within the tumor tissue promote immune evasion as a critical strategy for cancer development and progression, but the underlying mechanisms remain poorly understood. In this study, we explore the role of endothelial cells (ECs) in the regulation of the immunosuppressive tumor microenvironment. Using mouse pancreatic ductal adenocarcinoma (PDAC) models, we found that canonical Notch signaling in endothelial cells suppresses the recruitment of antitumor T cells and promotes tumor progression by inhibiting the pro-inflammatory functions of cancer-associated fibroblasts (CAFs). Abrogation of endothelial Notch signaling modulates EC-derived angiocrine factors to enhance the pro-inflammatory activities of CAFs, which drive CXCL9/10-CXCR3-mediated T cell recruitment to inhibit tumor growth. Additionally, abrogation of endothelial Notch unleashed interferon gamma responses in the tumor microenvironment, upregulated PDL1 expression on tumor cells, and sensitized PDAC to PD1-based immunotherapy. Collectively, these data uncover a pivotal role of endothelial cells in shaping the immunosuppressive microenvironment, and suggest the potential of targeting EC-CAF interaction as a novel therapeutic modality to boost antitumor immunity.

13.
J Virol ; 86(18): 9696-707, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740393

RESUMO

Genome-wide chromatin immunoprecipitation assays indicate that the promoter-proximal pausing of RNA polymerase II (RNAPII) is an important postinitiation step for gene regulation. During latent infection, the majority of Kaposi's sarcoma-associated herpesvirus (KSHV) genes is silenced via repressive histone marks on their promoters. Despite the absence of their expression during latency, however, several lytic promoters are enriched with activating histone marks, suggesting that mechanisms other than heterochromatin-mediated suppression contribute to preventing lytic gene expression. Here, we show that the RNAPII-mediated transcription of the KSHV OriLytL, K5, K6, and K7 (OriLytL-K7) lytic genes is paused at the elongation step during latency. Specifically, the RNAPII-mediated transcription is stalled by the host's negative elongation factor (NELF) at the promoter regions of OriLytL-K7 lytic genes during latency, leading to the hyperphosphorylation of the serine 5 residue and the hypophosphorylation of the serine 2 of the C-terminal domain of the RNAPII large subunit, a hallmark of stalled RNAPII. Consequently, depletion of NELF expression induced transition of stalled RNAPII into a productive transcription elongation at the promoter-proximal regions of OriLytL-K7 lytic genes, leading to their RTA-independent expression. Using an RTA-deficient recombinant KSHV, we also showed that expression of the K5, K6, and K7 lytic genes was highly inducible upon external stimuli compared to other lytic genes that lack RNAPII on their promoters during latency. These results indicate that the transcription elongation of KSHV OriLytL-K7 lytic genes is inhibited by NELF during latency, but can also be promptly reactivated in an RTA-independent manner upon external stimuli.


Assuntos
Herpesvirus Humano 8/fisiologia , RNA Polimerase II/fisiologia , Fatores de Transcrição/fisiologia , Sequência de Bases , Linhagem Celular , Regulação Viral da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Virais , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/fisiologia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Transativadores/antagonistas & inibidores , Transativadores/genética , Transativadores/fisiologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Latência Viral/genética , Latência Viral/fisiologia
14.
J Virol ; 86(18): 9708-20, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740391

RESUMO

Efficient genetic modification of herpesviruses such as Kaposi's sarcoma-associated herpesvirus (KSHV) has come to rely on bacterial artificial chromosome (BAC) technology. In order to facilitate this approach, we generated a new KSHV BAC clone, called BAC16, derived from the rKSHV.219 virus, which stems from KSHV and Epstein-Barr virus-coinfected JSC1 primary effusion lymphoma (PEL) cells. Restriction enzyme and complete sequencing data demonstrate that the KSHV of JSC1 PEL cells showed a minimal level of sequence variation across the entire viral genome compared to the complete genomic sequence of other KSHV strains. BAC16 not only stably propagated in both Escherichia coli and mammalian cells without apparent genetic rearrangements, but also was capable of robustly producing infectious virions (∼5 × 10(7)/ml). We also demonstrated the utility of BAC16 by generating deletion mutants of either the K3 or K5 genes, whose products are E3 ligases of the membrane-associated RING-CH (MARCH) family. While previous studies have shown that individual expression of either K3 or K5 results in efficient downregulation of the surface expression of major histocompatibility complex class I (MHC-I) molecules, we found that K5, but not K3, was the primary factor critical for the downregulation of MHC-I surface expression during KSHV lytic reactivation or following de novo infection. The data presented here demonstrate the utility of BAC16 for the generation and characterization of KSHV knockout and mutant recombinants and further emphasize the importance of functional analysis of viral genes in the context of the KSHV genome besides the study of individual gene expression.


Assuntos
Cromossomos Artificiais Bacterianos/genética , Herpesvirus Humano 8/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Chlorocebus aethiops , Clonagem Molecular , DNA Viral/genética , Escherichia coli/genética , Deleção de Genes , Regulação Viral da Expressão Gênica , Genoma Viral , Herpesvirus Humano 8/patogenicidade , Herpesvirus Humano 8/fisiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/fisiologia , Linfoma de Efusão Primária/virologia , Dados de Sequência Molecular , Mutação , Plasmídeos/genética , Células Vero , Proteínas Virais/genética , Proteínas Virais/fisiologia
15.
Nat Commun ; 14(1): 2307, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37085516

RESUMO

The intestinal lamina propria contains a diverse network of fibroblasts that provide key support functions to cells within their local environment. Despite this, our understanding of the diversity, location and ontogeny of fibroblasts within and along the length of the intestine remains incomplete. Here we show that the small and large intestinal lamina propria contain similar fibroblast subsets that locate in specific anatomical niches. Nevertheless, we find that the transcriptional profile of similar fibroblast subsets differs markedly between the small intestine and colon suggesting region specific functions. We perform in vivo transplantation and lineage-tracing experiments to demonstrate that adult intestinal fibroblast subsets, smooth muscle cells and pericytes derive from Gli1-expressing precursors present in embryonic day 12.5 intestine. Trajectory analysis of single cell RNA-seq datasets of E12.5 and adult mesenchymal cells suggest that adult smooth muscle cells and fibroblasts derive from distinct embryonic intermediates and that adult fibroblast subsets develop in a linear trajectory from CD81+ fibroblasts. Finally, we provide evidence that colonic subepithelial PDGFRαhi fibroblasts comprise several functionally distinct populations that originate from an Fgfr2-expressing fibroblast intermediate. Our results provide insights into intestinal stromal cell diversity, location, function, and ontogeny, with implications for intestinal development and homeostasis.


Assuntos
Intestino Grosso , Células-Tronco Mesenquimais , Colo , Fibroblastos/metabolismo , Intestino Grosso/anatomia & histologia , Intestino Grosso/citologia , Intestino Delgado , Intestinos/anatomia & histologia , Intestinos/citologia , Proteína GLI1 em Dedos de Zinco/genética , Células-Tronco Mesenquimais/metabolismo
16.
PLoS Pathog ; 6(7): e1001013, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20661424

RESUMO

Epigenetic modifications of the herpesviral genome play a key role in the transcriptional control of latent and lytic genes during a productive viral lifecycle. In this study, we describe for the first time a comprehensive genome-wide ChIP-on-Chip analysis of the chromatin associated with the Kaposi's sarcoma-associated herpesvirus (KSHV) genome during latency and lytic reactivation. Depending on the gene expression class, different combinations of activating [acetylated H3 (AcH3) and H3K4me3] and repressive [H3K9me3 and H3K27me3] histone modifications are associated with the viral latent genome, which changes upon reactivation in a manner that is correlated with their expression. Specifically, both the activating marks co-localize on the KSHV latent genome, as do the repressive marks. However, the activating and repressive histone modifications are mutually exclusive of each other on the bulk of the latent KSHV genome. The genomic region encoding the IE genes ORF50 and ORF48 possesses the features of a bivalent chromatin structure characterized by the concomitant presence of the activating H3K4me3 and the repressive H3K27me3 marks during latency, which rapidly changes upon reactivation with increasing AcH3 and H3K4me3 marks and decreasing H3K27me3. Furthermore, EZH2, the H3K27me3 histone methyltransferase of the Polycomb group proteins (PcG), colocalizes with the H3K27me3 mark on the entire KSHV genome during latency, whereas RTA-mediated reactivation induces EZH2 dissociation from the genomic regions encoding IE and E genes concurrent with decreasing H3K27me3 level and increasing IE/E lytic gene expression. Moreover, either the inhibition of EZH2 expression by a small molecule inhibitor DZNep and RNAi knockdown, or the expression of H3K27me3-specific histone demethylases apparently induced the KSHV lytic gene expression cascade. These data indicate that histone modifications associated with the KSHV latent genome are involved in the regulation of latency and ultimately in the control of the temporal and sequential expression of the lytic gene cascade. In addition, the PcG proteins play a critical role in the control of KSHV latency by maintaining a reversible heterochromatin on the KSHV lytic genes. Thus, the regulation of the spatial and temporal association of the PcG proteins with the KSHV genome may be crucial for propagating the KSHV lifecycle.


Assuntos
Cromatina/genética , Epigênese Genética , Genoma Viral , Herpesvirus Humano 8/genética , Latência Viral/genética , Linhagem Celular , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/fisiologia , Histonas/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética
17.
Sci Immunol ; 5(47)2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32404436

RESUMO

Gastrointestinal symptoms and fecal shedding of SARS-CoV-2 RNA are frequently observed in COVID-19 patients. However, it is unclear whether SARS-CoV-2 replicates in the human intestine and contributes to possible fecal-oral transmission. Here, we report productive infection of SARS-CoV-2 in ACE2+ mature enterocytes in human small intestinal enteroids. Expression of two mucosa-specific serine proteases, TMPRSS2 and TMPRSS4, facilitated SARS-CoV-2 spike fusogenic activity and promoted virus entry into host cells. We also demonstrate that viruses released into the intestinal lumen were inactivated by simulated human colonic fluid, and infectious virus was not recovered from the stool specimens of COVID-19 patients. Our results highlight the intestine as a potential site of SARS-CoV-2 replication, which may contribute to local and systemic illness and overall disease progression.


Assuntos
Betacoronavirus/fisiologia , Enterócitos/virologia , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Internalização do Vírus , Enzima de Conversão de Angiotensina 2 , Animais , Linhagem Celular , Duodeno/citologia , Enterócitos/patologia , Humanos , Camundongos , Organoides/virologia , Peptidil Dipeptidase A/metabolismo , Rotavirus/fisiologia , SARS-CoV-2 , Vesiculovirus/genética
18.
Nat Commun ; 9(1): 1485, 2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29662124

RESUMO

Cohesin is a multi-subunit nuclear protein complex that coordinates sister chromatid separation during cell division. Highly frequent somatic mutations in genes encoding core cohesin subunits have been reported in multiple cancer types. Here, using a genome-wide CRISPR-Cas9 screening approach to identify host dependency factors and novel innate immune regulators of rotavirus (RV) infection, we demonstrate that the loss of STAG2, an important component of the cohesin complex, confers resistance to RV replication in cell culture and human intestinal enteroids. Mechanistically, STAG2 deficiency results in spontaneous genomic DNA damage and robust interferon (IFN) expression via the cGAS-STING cytosolic DNA-sensing pathway. The resultant activation of JAK-STAT signaling and IFN-stimulated gene (ISG) expression broadly protects against virus infections, including RVs. Our work highlights a previously undocumented role of the cohesin complex in regulating IFN homeostasis and identifies new therapeutic avenues for manipulating the innate immunity.


Assuntos
Antígenos Nucleares/imunologia , Proteínas de Ciclo Celular/imunologia , Proteínas Cromossômicas não Histona/imunologia , Interações Hospedeiro-Patógeno , Proteínas de Membrana/imunologia , Nucleotidiltransferases/imunologia , Rotavirus/imunologia , Esferoides Celulares/imunologia , Antígenos Nucleares/genética , Sistemas CRISPR-Cas , Células CACO-2 , Proteínas de Ciclo Celular/genética , Núcleo Celular/imunologia , Núcleo Celular/virologia , Proteínas Cromossômicas não Histona/genética , Dano ao DNA , Deleção de Genes , Edição de Genes , Regulação da Expressão Gênica , Genoma Humano , Células HEK293 , Células HT29 , Células HeLa , Humanos , Interferons/genética , Interferons/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia , Janus Quinases/genética , Janus Quinases/imunologia , Proteínas de Membrana/genética , Nucleotidiltransferases/genética , Rotavirus/crescimento & desenvolvimento , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Esferoides Celulares/virologia , Coesinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA