Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(3): 113888, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38416644

ABSTRACT

Higher-order genome structure influences the transcriptional regulation of cellular genes through the juxtaposition of regulatory elements, such as enhancers, close to promoters of target genes. While enhancer activation has emerged as an important facet of Kaposi sarcoma-associated herpesvirus (KSHV) biology, the mechanisms controlling enhancer-target gene expression remain obscure. Here, we discover that the KSHV genome tethering protein latency-associated nuclear antigen (LANA) potentiates enhancer-target gene expression in primary effusion lymphoma (PEL), a highly aggressive B cell lymphoma causally associated with KSHV. Genome-wide analyses demonstrate increased levels of enhancer RNA transcription as well as activating chromatin marks at LANA-bound enhancers. 3D genome conformation analyses identified genes critical for latency and tumorigenesis as targets of LANA-occupied enhancers, and LANA depletion results in their downregulation. These findings reveal a mechanism in enhancer-gene coordination and describe a role through which the main KSHV tethering protein regulates essential gene expression in PEL.


Subject(s)
Herpesvirus 8, Human , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Genome-Wide Association Study , Antigens, Viral/genetics , Antigens, Viral/metabolism , Promoter Regions, Genetic/genetics , Gene Expression Regulation , Virus Latency
2.
bioRxiv ; 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38293013

ABSTRACT

5-fluorouracil (5-FU) has been used for chemotherapy for colorectal and other cancers for over 50 years. The prevailing view of its mechanism of action is inhibition of thymidine synthase leading to defects in DNA replication and repair. However, 5-FU is also incorporated into RNA causing toxicity due to defects in RNA metabolism, inhibition of pseudouridine modification, and altered ribosome function. Here, we examine the impact of 5-FU on the expression and export of small RNAs (sRNAs) into small extracellular vesicles (sEVs). Moreover, we assess the role of 5-FU in regulation of post-transcriptional sRNA modifications (PTxM) using mass spectrometry approaches. EVs are secreted by all cells and contain a variety of proteins and RNAs that can function in cell-cell communication. PTxMs on cellular and extracellular sRNAs provide yet another layer of gene regulation. We found that treatment of the colorectal cancer (CRC) cell line DLD-1 with 5-FU led to surprising differential export of miRNA snRNA, and snoRNA transcripts. Strikingly, 5-FU treatment significantly decreased the levels of pseudouridine on both cellular and secreted EV sRNAs. In contrast, 5-FU exposure led to increased levels of cellular sRNAs containing a variety of methyl-modified bases. Our results suggest that 5-FU exposure leads to altered expression, base modifications, and mislocalization of EV base-modified sRNAs.

3.
J Med Virol ; 96(2): e29424, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38285432

ABSTRACT

Protein kinase R (PKR) is a double-stranded RNA (dsRNA) binding protein that plays a crucial role in innate immunity during viral infection and can restrict both DNA and RNA viruses. The potency of its antiviral function is further reflected by the large number of viral-encoded PKR antagonists. However, much about the regulation of dsRNA accumulation and PKR activation during viral infection remains unknown. Since DNA viruses do not have an RNA genome or RNA replication intermediates like RNA viruses do, PKR-mediated dsRNA detection in the context of DNA virus infection is particularly intriguing. Here, we review the current state of knowledge regarding the regulation of PKR activation and its antagonism during infection with DNA viruses.


Subject(s)
DNA Virus Infections , Protein Kinases , RNA , Humans , Immunity, Innate
4.
Cell Rep ; 42(8): 112928, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37542720

ABSTRACT

Identifying molecular circuits that control adipose tissue macrophage (ATM) function is necessary to understand how ATMs contribute to tissue homeostasis and obesity-induced insulin resistance. In this study, we find that mice with a myeloid-specific knockout of the miR-23-27-24 clusters of microRNAs (miRNAs) gain less weight on a high-fat diet but exhibit worsened glucose and insulin tolerance. Analysis of ATMs from these mice shows selectively reduced numbers and proliferation of a recently reported subset of lipid-associated CD9+Trem2+ ATMs (lipid-associated macrophages [LAMs]). Leveraging the role of miRNAs to control networks of genes, we use RNA sequencing (RNA-seq), functional screens, and biochemical assays to identify candidate target transcripts that regulate proliferation-associated signaling. We determine that miR-23 directly targets the mRNA of Eif4ebp2, a gene that restricts protein synthesis and proliferation in macrophages. Altogether, our study demonstrates that control of proliferation of a protective subset of LAMs by noncoding RNAs contributes to protection against diet-induced obesity metabolic dysfunction.


Subject(s)
Insulin Resistance , MicroRNAs , Mice , Animals , Adipose Tissue/metabolism , Obesity/genetics , Obesity/metabolism , Macrophages/metabolism , Insulin Resistance/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Diet, High-Fat , Lipids , Cell Proliferation , Mice, Inbred C57BL , Inflammation/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism
5.
J Immunol ; 211(4): 563-575, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37341499

ABSTRACT

Activated T cells undergo metabolic reprogramming to meet anabolic, differentiation, and functional demands. Glutamine supports many processes in activated T cells, and inhibition of glutamine metabolism alters T cell function in autoimmune disease and cancer. Multiple glutamine-targeting molecules are under investigation, yet the precise mechanisms of glutamine-dependent CD8 T cell differentiation remain unclear. We show that distinct strategies of glutamine inhibition by glutaminase-specific inhibition with small molecule CB-839, pan-glutamine inhibition with 6-diazo-5-oxo-l-norleucine (DON), or by glutamine-depleted conditions (No Q) produce distinct metabolic differentiation trajectories in murine CD8 T cells. T cell activation with CB-839 treatment had a milder effect than did DON or No Q treatment. A key difference was that CB-839-treated cells compensated with increased glycolytic metabolism, whereas DON and No Q-treated cells increased oxidative metabolism. However, all glutamine treatment strategies elevated CD8 T cell dependence on glucose metabolism, and No Q treatment caused adaptation toward reduced glutamine dependence. DON treatment reduced histone modifications and numbers of persisting cells in adoptive transfer studies, but those T cells that remained could expand normally upon secondary Ag encounter. In contrast, No Q-treated cells persisted well yet demonstrated decreased secondary expansion. Consistent with reduced persistence, CD8 T cells activated in the presence of DON had reduced ability to control tumor growth and reduced tumor infiltration in adoptive cell therapy. Overall, each approach to inhibit glutamine metabolism confers distinct effects on CD8 T cells and highlights that targeting the same pathway in different ways can elicit opposing metabolic and functional outcomes.


Subject(s)
Diazooxonorleucine , Neoplasms , Animals , Mice , Diazooxonorleucine/pharmacology , Glutamine/metabolism , Neoplasms/therapy , Neoplasms/metabolism , CD8-Positive T-Lymphocytes/metabolism
6.
Nat Commun ; 14(1): 1367, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36914661

ABSTRACT

Adenosine-to-inosine RNA editing is a major contributor to transcriptome diversity in animals with far-reaching biological consequences. Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of several human malignancies including primary effusion lymphoma (PEL). The extent of RNA editing within the KSHV transcriptome is unclear as is its contribution to the viral lifecycle. Here, we leverage a combination of biochemical and genomic approaches to determine the RNA editing landscape in host- and KSHV transcriptomes during both latent and lytic replication in PEL. Analysis of RNA editomes reveals it is dynamic, with increased editing upon reactivation and the potential to deregulate pathways critical for latency and tumorigenesis. In addition, we identify conserved RNA editing events within a viral microRNA and discover their role in miRNA biogenesis as well as viral infection. Together, these results describe the editome of PEL cells as well as a critical role for A-to-I editing in the KSHV lifecycle.


Subject(s)
Herpesvirus 8, Human , Lymphoma, Primary Effusion , MicroRNAs , Sarcoma, Kaposi , Animals , Humans , Herpesvirus 8, Human/metabolism , Virus Latency/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Virus Replication/genetics , Gene Expression Regulation, Viral
7.
Sci Rep ; 12(1): 19101, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36351985

ABSTRACT

Natural infection as well as vaccination with live or attenuated viruses elicit tissue resident, CD8+ memory T cell (Trm) response. Trm cells so elicited act quickly upon reencounter with the priming agent to protect the host. These Trm cells express a unique molecular signature driven by the master regulators-Runx3 and Hobit. We previously reported that intranasal instillation of a subunit vaccine in a prime boost vaccination regimen installed quick-acting, CD8+ Trm cells in the lungs that protected against lethal vaccinia virus challenge. It remains unexplored whether CD8+ Trm responses so elicited are driven by a similar molecular signature as those elicited by microbes in a real infection or by live, attenuated pathogens in conventional vaccination. We found that distinct molecular signatures distinguished subunit vaccine-elicited lung interstitial CD8+ Trm cells from subunit vaccine-elicited CD8+ effector memory and splenic memory T cells. Nonetheless, the transcriptome signature of subunit vaccine elicited CD8+ Trm resembled those elicited by virus infection or vaccination. Clues to the basis of tissue residence and function of vaccine specific CD8+ Trm cells were found in transcripts that code for chemokines and chemokine receptors, purinergic receptors, and adhesins when compared to CD8+ effector and splenic memory T cells. Our findings inform the utility of protein-based subunit vaccination for installing CD8+ Trm cells in the lungs to protect against respiratory infectious diseases that plague humankind.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Vaccination , Lung , Vaccines, Subunit
8.
Eur J Cell Biol ; 101(3): 151239, 2022.
Article in English | MEDLINE | ID: mdl-35623231

ABSTRACT

Approximately 170 RNA modifications have been identified and these are critical for determining the fate and function of cellular RNAs. Similar to human transcripts, viral RNAs possess an extensive RNA modification landscape. While initial efforts largely focused on investigating the RNA modification landscape in the context of RNA virus infection, a growing body of work has explored the impact of RNA modifications on DNA virus biology. These studies have revealed roles for RNA modifications in DNA virus infection, including gene regulation and viral pathogenesis. In this review, we will discuss the current knowledge on how RNA modifications impact DNA virus biology.


Subject(s)
DNA Virus Infections , Virus Replication , Biology , DNA Virus Infections/genetics , Gene Expression Regulation , Humans , RNA, Viral/genetics
9.
Mol Cancer ; 21(1): 74, 2022 03 12.
Article in English | MEDLINE | ID: mdl-35279145

ABSTRACT

BACKGROUND: Epithelial-to-mesenchymal transition (EMT) is a process linked to metastasis and drug resistance with non-coding RNAs (ncRNAs) playing pivotal roles. We previously showed that miR-100 and miR-125b, embedded within the third intron of the ncRNA host gene MIR100HG, confer resistance to cetuximab, an anti-epidermal growth factor receptor (EGFR) monoclonal antibody, in colorectal cancer (CRC). However, whether the MIR100HG transcript itself has a role in cetuximab resistance or EMT is unknown. METHODS: The correlation between MIR100HG and EMT was analyzed by curating public CRC data repositories. The biological roles of MIR100HG in EMT, metastasis and cetuximab resistance in CRC were determined both in vitro and in vivo. The expression patterns of MIR100HG, hnRNPA2B1 and TCF7L2 in CRC specimens from patients who progressed on cetuximab and patients with metastatic disease were analyzed by RNAscope and immunohistochemical staining. RESULTS: The expression of MIR100HG was strongly correlated with EMT markers and acted as a positive regulator of EMT. MIR100HG sustained cetuximab resistance and facilitated invasion and metastasis in CRC cells both in vitro and in vivo. hnRNPA2B1 was identified as a binding partner of MIR100HG. Mechanistically, MIR100HG maintained mRNA stability of TCF7L2, a major transcriptional coactivator of the Wnt/ß-catenin signaling, by interacting with hnRNPA2B1. hnRNPA2B1 recognized the N6-methyladenosine (m6A) site of TCF7L2 mRNA in the presence of MIR100HG. TCF7L2, in turn, activated MIR100HG transcription, forming a feed forward regulatory loop. The MIR100HG/hnRNPA2B1/TCF7L2 axis was augmented in specimens from CRC patients who either developed local or distant metastasis or had disease progression that was associated with cetuximab resistance. CONCLUSIONS: MIR100HG and hnRNPA2B1 interact to control the transcriptional activity of Wnt signaling in CRC via regulation of TCF7L2 mRNA stability. Our findings identified MIR100HG as a potent EMT inducer in CRC that may contribute to cetuximab resistance and metastasis by activation of a MIR100HG/hnRNPA2B1/TCF7L2 feedback loop.


Subject(s)
Colorectal Neoplasms , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , MicroRNAs , RNA, Long Noncoding , Cell Line, Tumor , Cell Movement/genetics , Cetuximab/genetics , Cetuximab/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Wnt Signaling Pathway/genetics
10.
Immunity ; 55(1): 65-81.e9, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34767747

ABSTRACT

Antigenic stimulation promotes T cell metabolic reprogramming to meet increased biosynthetic, bioenergetic, and signaling demands. We show that the one-carbon (1C) metabolism enzyme methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) regulates de novo purine synthesis and signaling in activated T cells to promote proliferation and inflammatory cytokine production. In pathogenic T helper-17 (Th17) cells, MTHFD2 prevented aberrant upregulation of the transcription factor FoxP3 along with inappropriate gain of suppressive capacity. MTHFD2 deficiency also promoted regulatory T (Treg) cell differentiation. Mechanistically, MTHFD2 inhibition led to depletion of purine pools, accumulation of purine biosynthetic intermediates, and decreased nutrient sensor mTORC1 signaling. MTHFD2 was also critical to regulate DNA and histone methylation in Th17 cells. Importantly, MTHFD2 deficiency reduced disease severity in multiple in vivo inflammatory disease models. MTHFD2 is thus a metabolic checkpoint to integrate purine metabolism with pathogenic effector cell signaling and is a potential therapeutic target within 1C metabolism pathways.


Subject(s)
Inflammation/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Purines/biosynthesis , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cell Differentiation , Cytokines/metabolism , DNA Methylation , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mice, Transgenic , Mutation/genetics , Signal Transduction
11.
Heliyon ; 7(12): e08519, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34934837

ABSTRACT

Extracellular vesicles (EVs) are capable of transferring cargo from donor to recipient cells, but precisely how cargo content is regulated for export is mostly unknown. For miRNA cargo, we previously showed that when compared to isogenic colorectal cancer (CRC) cells expressing wild-type KRAS, a distinct subset of miRNAs are differentially enriched in EVs from KRAS mutant active CRC cells, with miR-100 being one of the most enriched. The mechanisms that could explain how miR-100 and other miRNAs are differentially exported into EVs have not been fully elucidated. Here, we tested the effect of N6-methyladenosine (m6A) modification on miRNA export into EVs by depletion of METTL3 and ALKBH5, a writer and eraser of m6A modification, respectively. While the effects of ALKBH5 knockdown were quite modest, decreased levels of METTL3 led to reduced cellular and extracellular levels of a subset of miRNAs that contain consensus sequences for m6A modification. Functional testing of EVs prepared from cells expressing shRNAs against METTL3 showed that they were less capable of conferring colony growth in 3D to wild-type KRAS cells and were also largely incapable of conferring the spread of cetuximab resistance. Our data support a role for METTL3 modification on cellular miRNA levels and export of specific miRNAs.

12.
Viruses ; 13(6)2021 06 08.
Article in English | MEDLINE | ID: mdl-34201386

ABSTRACT

Packaging of segmented, double-stranded RNA viral genomes requires coordination of viral proteins and RNA segments. For mammalian orthoreovirus (reovirus), evidence suggests either all ten or zero viral RNA segments are simultaneously packaged in a highly coordinated process hypothesized to exclude host RNA. Accordingly, reovirus generates genome-containing virions and "genomeless" top component particles. Whether reovirus virions or top component particles package host RNA is unknown. To gain insight into reovirus packaging potential and mechanisms, we employed next-generation RNA-sequencing to define the RNA content of enriched reovirus particles. Reovirus virions exclusively packaged viral double-stranded RNA. In contrast, reovirus top component particles contained similar proportions but reduced amounts of viral double-stranded RNA and were selectively enriched for numerous host RNA species, especially short, non-polyadenylated transcripts. Host RNA selection was not dependent on RNA abundance in the cell, and specifically enriched host RNAs varied for two reovirus strains and were not selected solely by the viral RNA polymerase. Collectively, these findings indicate that genome packaging into reovirus virions is exquisitely selective, while incorporation of host RNAs into top component particles is differentially selective and may contribute to or result from inefficient viral RNA packaging.


Subject(s)
Host Microbial Interactions/genetics , RNA, Double-Stranded/metabolism , Reoviridae/genetics , Viral Genome Packaging/genetics , Virion/genetics , Virion/physiology , Animals , Cell Line , Genome, Viral , Mice , RNA-Seq , Reoviridae/physiology , Viral Genome Packaging/physiology , Viral Proteins/genetics , Viral Proteins/metabolism
13.
Cell Rep ; 35(2): 108976, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852834

ABSTRACT

RIG-I-like receptors (RLRs) are involved in the discrimination of self versus non-self via the recognition of double-stranded RNA (dsRNA). Emerging evidence suggests that immunostimulatory dsRNAs are ubiquitously expressed but are disrupted or sequestered by cellular RNA binding proteins (RBPs). TDP-43 is an RBP associated with multiple neurological disorders and is essential for cell viability. Here, we demonstrate that TDP-43 regulates the accumulation of immunostimulatory dsRNA. The immunostimulatory RNA is identified as RNA polymerase III transcripts, including 7SL and Alu retrotransposons, and we demonstrate that the RNA-binding activity of TDP-43 is required to prevent immune stimulation. The dsRNAs activate a RIG-I-dependent interferon (IFN) response, which promotes necroptosis. Genetic inactivation of the RLR-pathway rescues the interferon-mediated cell death associated with loss of TDP-43. Collectively, our study describes a role for TDP-43 in preventing the accumulation of endogenous immunostimulatory dsRNAs and uncovers an intricate relationship between the control of cellular gene expression and IFN-mediated cell death.


Subject(s)
DEAD Box Protein 58/genetics , DNA-Binding Proteins/genetics , Herpesvirus 8, Human/genetics , Necroptosis/genetics , RNA, Double-Stranded/genetics , Receptors, Immunologic/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Alu Elements , Cell Line, Tumor , Cell Survival , Cytokines/genetics , Cytokines/immunology , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Regulation , HEK293 Cells , Herpesvirus 8, Human/growth & development , Herpesvirus 8, Human/immunology , Humans , Immunization , Interferons/genetics , Interferons/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Necroptosis/immunology , Neurons/immunology , Neurons/virology , RNA Polymerase III/genetics , RNA Polymerase III/immunology , RNA, Double-Stranded/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/immunology , RNA, Viral/genetics , RNA, Viral/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Signal Recognition Particle/genetics , Signal Recognition Particle/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Ubiquitins/genetics , Ubiquitins/immunology
14.
Nat Commun ; 11(1): 3345, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620802

ABSTRACT

Nonsense-mediated mRNA decay (NMD) is an evolutionarily conserved RNA decay mechanism that has emerged as a potent cell-intrinsic restriction mechanism of retroviruses and positive-strand RNA viruses. However, whether NMD is capable of restricting DNA viruses is not known. The DNA virus Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma and primary effusion lymphoma (PEL). Here, we demonstrate that NMD restricts KSHV lytic reactivation. Leveraging high-throughput transcriptomics we identify NMD targets transcriptome-wide in PEL cells and identify host and viral RNAs as substrates. Moreover, we identified an NMD-regulated link between activation of the unfolded protein response and transcriptional activation of the main KSHV transcription factor RTA, itself an NMD target. Collectively, our study describes an intricate relationship between cellular targets of an RNA quality control pathway and KSHV lytic gene expression, and demonstrates that NMD can function as a cell intrinsic restriction mechanism acting upon DNA viruses.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Nonsense Mediated mRNA Decay , RNA, Viral/metabolism , Virus Activation/genetics , Cell Line, Tumor , HEK293 Cells , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , RNA, Messenger/metabolism , RNA-Seq , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Activation , Unfolded Protein Response/genetics , Virus Latency/genetics
15.
JCI Insight ; 5(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32493845

ABSTRACT

Recently, we reported that expression of endogenous retroviruses (ERVs) is associated with response to immune checkpoint blockade (ICB) in renal cell carcinoma (RCC). We show that decitabine, a DNA hypomethylating agent, activates transposable element (TE) expression (LINE1 and ERVs ERV3-2 and ERV4700) and antiviral signaling to potentially enhance response to ICB in kidney cancer cell lines and primary cells. KO of RIGI and MDA5 dsRNA sensors attenuated activation of antiviral signaling associated with DNA hypomethylation, and RIGI and MDA5 IPs showed increased ERV binding with decitabine treatment. Bioinformatic analyses showed the decitabine-induced signature could be associated with increased immune infiltration and response to ICB. Cytokine secretion induced by decitabine could modestly improve T cell activation and robustly enhanced T cell migration. In a small retrospective cohort of metastatic clear cell RCC (ccRCC) patients treated with anti-PD1/PDL1 blockade, activation of some antiviral genes was significantly higher in responders. Thus, we identified a potential strategy to induce TE expression through inhibition of DNA methylation in modulating T cell action via regulation of the innate antiviral pathway.


Subject(s)
Carcinoma, Renal Cell/immunology , DNA Methylation , DNA Transposable Elements/immunology , DNA, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/immunology , Kidney Neoplasms/immunology , Signal Transduction/immunology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Humans , Kidney Neoplasms/pathology
16.
J Virol ; 93(23)2019 12 01.
Article in English | MEDLINE | ID: mdl-31511389

ABSTRACT

The RIG-I-like receptors (RLRs) are double-stranded RNA-binding proteins that play a role in initiating and modulating cell intrinsic immunity through the recognition of RNA features typically absent from the host transcriptome. While they are initially characterized in the context of RNA virus infection, evidence has now accumulated establishing the role of RLRs in DNA virus infection. Here, we review recent advances in the RLR-mediated restriction of DNA virus infection with an emphasis on the RLR ligands sensed.


Subject(s)
DEAD Box Protein 58/metabolism , DNA Virus Infections/metabolism , RNA-Binding Proteins/metabolism , Sensory Receptor Cells/metabolism , Humans , Immunity, Innate , RNA Virus Infections , Receptors, Immunologic , Transcriptome
17.
PLoS Pathog ; 15(6): e1007852, 2019 06.
Article in English | MEDLINE | ID: mdl-31188901

ABSTRACT

Precise promoter annotation is required for understanding the mechanistic basis of transcription initiation. In the context of complex genomes, such as herpesviruses where there is extensive genic overlap, identification of transcription start sites (TSSs) is particularly problematic and cannot be comprehensively accessed by standard RNA sequencing approaches. Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic gammaherpesvirus and the etiological agent of Kaposi's sarcoma and the B cell lymphoma primary effusion lymphoma (PEL). Here, we leverage RNA annotation and mapping of promoters for analysis of gene expression (RAMPAGE) and define KSHV TSSs transcriptome-wide and at nucleotide resolution in two widely used models of KSHV infection, namely iSLK.219 cells and the PEL cell line TREx-BCBL1-RTA. By mapping TSSs over a 96 h time course of reactivation we confirm 48 of 50 previously identified TSSs. Moreover, we identify over 100 novel transcription start site clusters (TSCs) in each cell line. Our analyses identified cell-type specific differences in TSC positions as well as promoter strength, and defined motifs within viral core promoters. Collectively, by defining TSSs at high resolution we have greatly expanded the transcriptional landscape of the KSHV genome and identified transcriptional control mechanisms at play during KSHV lytic reactivation.


Subject(s)
Genome, Viral/physiology , Herpesviridae Infections/metabolism , Herpesvirus 8, Human/physiology , Transcription Initiation, Genetic/physiology , Virus Activation , HEK293 Cells , Herpesviridae Infections/virology , Humans
18.
J Autoimmun ; 100: 40-51, 2019 06.
Article in English | MEDLINE | ID: mdl-30826177

ABSTRACT

Various sensors that detect double-stranded RNA, presumably of viral origin, exist in eukaryotic cells and induce IFN-responses. Ongoing IFN-responses have also been documented in a variety of human autoimmune diseases including relapsing-remitting multiple sclerosis (RRMS) but their origins remain obscure. We find increased IFN-responses in leukocytes in relapsing-remitting multiple sclerosis at distinct stages of disease. Moreover, endogenous RNAs isolated from blood cells of these same patients recapitulate this IFN-response if transfected into naïve cells. These endogenous RNAs are double-stranded RNAs, contain Alu and Line elements and are transcribed from leukocyte transcriptional enhancers. Thus, transcribed endogenous retrotransposon elements can co-opt pattern recognition sensors to induce IFN-responses in RRMS.


Subject(s)
Alu Elements/immunology , Interferons/immunology , Long Interspersed Nucleotide Elements/immunology , Multiple Sclerosis/immunology , RNA, Double-Stranded/immunology , Adult , Aged , Female , Humans , Male , Middle Aged , Multiple Sclerosis/pathology
19.
Nat Commun ; 9(1): 4841, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451863

ABSTRACT

The RIG-I like receptors (RLRs) RIG-I and MDA5 are cytosolic RNA helicases best characterized as restriction factors for RNA viruses. However, evidence suggests RLRs participate in innate immune recognition of other pathogens, including DNA viruses. Kaposi's sarcoma-associated herpesvirus (KSHV) is a human gammaherpesvirus and the etiological agent of Kaposi's sarcoma and primary effusion lymphoma (PEL). Here, we demonstrate that RLRs restrict KSHV lytic reactivation and we demonstrate that restriction is facilitated by the recognition of host-derived RNAs. Misprocessed noncoding RNAs represent an abundant class of RIG-I substrates, and biochemical characterizations reveal that an infection-dependent reduction in the cellular triphosphatase DUSP11 results in an accumulation of select triphosphorylated noncoding RNAs, enabling their recognition by RIG-I. These findings reveal an intricate relationship between RNA processing and innate immunity, and demonstrate that an antiviral innate immune response can be elicited by the sensing of misprocessed cellular RNAs.


Subject(s)
DEAD Box Protein 58/genetics , Herpesvirus 8, Human/immunology , Host-Pathogen Interactions , Interferon-Induced Helicase, IFIH1/genetics , RNA Processing, Post-Transcriptional , RNA, Untranslated/genetics , Base Sequence , Cell Line, Tumor , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/immunology , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/immunology , Gene Expression Profiling , HEK293 Cells , Herpesvirus 8, Human/genetics , Humans , Immunity, Innate , Interferon-Induced Helicase, IFIH1/antagonists & inhibitors , Interferon-Induced Helicase, IFIH1/immunology , Lymphocytes/immunology , Lymphocytes/virology , Nucleic Acid Conformation , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Untranslated/immunology , Receptors, Immunologic , Signal Transduction , Virus Activation
20.
Viruses ; 10(7)2018 07 06.
Article in English | MEDLINE | ID: mdl-29986386

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human gammaherpesvirus and the etiological agent of Kaposi's sarcoma. KSHV is also causally associated with the development of lymphoproliferative diseases, including primary effusion lymphoma (PEL). KSHV reactivation from latency plays an integral role in the progression to KSHV-associated disease as several lytic proteins have angiogenic and anti-apoptotic functions essential to the tumor microenvironment. Thus, restriction of KSHV reactivation represents an attractive therapeutic target. Here, we demonstrate that the cellular protein Fused-in-sarcoma (FUS) restricts KSHV lytic reactivation in PEL and in an epithelial cell-based model. Depletion of FUS significantly enhances viral mRNA and protein expression, resulting in increased viral replication and production of infectious virions. Chromatin immunoprecipitation analyses demonstrate that FUS is present at several KSHV lytic cycle genes during the latent stage of infection. We further demonstrate that FUS interacts with RNA polymerase II and negatively affects Serine-2 phosphorylation of its C-terminal domain at the KSHV RTA gene, decreasing nascent RNA synthesis. Knockdown of FUS increases transcription of RTA, thus driving enhanced expression of KSHV lytic genes. Collectively, these data reveal a novel role for FUS in regulating viral gene expression and are the first to demonstrate its role as a viral restriction factor.


Subject(s)
Gene Expression Regulation, Viral , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Herpesvirus 8, Human/genetics , RNA-Binding Protein FUS/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Knockdown Techniques , Humans , Phosphorylation , Protein Transport , RNA Polymerase II/metabolism , RNA, Small Interfering/genetics , RNA-Binding Protein FUS/genetics , Virus Activation , Virus Latency/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...