Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
J Virol ; 98(6): e0017924, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38695538

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) belongs to the gamma-herpesvirus family and is a well-known human oncogenic virus. In infected cells, the viral genome of 165 kbp is circular DNA wrapped in chromatin. The tight control of gene expression is critical for latency, the transition into the lytic phase, and the development of viral-associated malignancies. Distal cis-regulatory elements, such as enhancers and silencers, can regulate gene expression in a position- and orientation-independent manner. Open chromatin is another characteristic feature of enhancers. To systematically search for enhancers, we cloned all the open chromatin regions in the KSHV genome downstream of the luciferase gene and tested their enhancer activity in infected and uninfected cells. A silencer was detected upstream of the latency-associated nuclear antigen promoter. Two constitutive enhancers were identified in the K12p-OriLyt-R and ORF29 Intron regions, where ORF29 Intron is a tissue-specific enhancer. The following promoters: OriLyt-L, PANp, ALTp, and the terminal repeats (TRs) acted as lytically induced enhancers. The expression of the replication and transcription activator (RTA), the master regulator of the lytic cycle, was sufficient to induce the activity of lytic enhancers in uninfected cells. We propose that the TRs that span about 24 kbp region serve as a "viral super-enhancer" that integrates the repressive effect of the latency-associated nuclear antigen (LANA) with the activating effect of RTA. Utilizing CRISPR activation and interference techniques, we determined the connections between these enhancers and their regulated genes. The silencer and enhancers described here provide an additional layer to the complex gene regulation of herpesviruses.IMPORTANCEIn this study, we performed a systematic functional assay to identify cis-regulatory elements within the genome of the oncogenic herpesvirus, Kaposi's sarcoma-associated herpesvirus (KSHV). Similar to other herpesviruses, KSHV presents both latent and lytic phases. Therefore, our assays were performed in uninfected cells, during latent infection, and under lytic conditions. We identified two constitutive enhancers, one of which seems to be a tissue-specific enhancer. In addition, four lytically induced enhancers, which are all responsive to the replication and transcription activator (RTA), were identified. Furthermore, a silencer was identified between the major latency promoter and the lytic gene locus. Utilizing CRISPR activation and interference techniques, we determined the connections between these enhancers and their regulated genes. The terminal repeats, spanning a region of about 24 kbp, seem like a "viral super-enhancer" that integrates the repressive effect of the latency-associated nuclear antigen (LANA) with the activating effect of RTA to regulate latency to lytic transition.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Viral , Genome, Viral , Herpesvirus 8, Human , Promoter Regions, Genetic , Virus Activation , Virus Latency , Humans , Antigens, Viral/genetics , Antigens, Viral/metabolism , Chromatin/metabolism , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Viral/genetics , Genome, Viral/genetics , HEK293 Cells , Herpesvirus 8, Human/genetics , Immediate-Early Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Terminal Repeat Sequences/genetics , Trans-Activators/metabolism , Virus Activation/genetics , Virus Latency/genetics
2.
Cell Host Microbe ; 32(5): 755-767.e4, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38653242

ABSTRACT

Kaposi sarcoma (KS) is the most common cancer in persons living with HIV. It is caused by KS-associated herpesvirus (KSHV). There exists no animal model for KS. Pronuclear injection of the 170,000-bp viral genome induces early-onset, aggressive angiosarcoma in transgenic mice. The tumors are histopathologically indistinguishable from human KS. As in human KS, all tumor cells express the viral latency-associated nuclear antigen (LANA). The tumors transcribe most viral genes, whereas endothelial cells in other organs only transcribe the viral latent genes. The tumor cells are of endothelial lineage and exhibit the same molecular pattern of pathway activation as KS, namely phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR, interleukin-10 (IL-10), and vascular endothelial growth factor (VEGF). The KSHV-induced tumors are more aggressive than Ha-ras-induced angiosarcomas. Overall survival is increased by prophylactic ganciclovir. Thus, whole-virus KSHV-transgenic mice represent an accurate model for KS and open the door for the genetic dissection of KS pathogenesis and evaluation of therapies, including vaccines.


Subject(s)
Disease Models, Animal , Hemangiosarcoma , Herpesvirus 8, Human , Mice, Transgenic , Sarcoma, Kaposi , Animals , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Mice , Hemangiosarcoma/virology , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Sarcoma, Kaposi/virology , Sarcoma, Kaposi/pathology , Genome, Viral , Humans , Antigens, Viral/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Ganciclovir/therapeutic use , Ganciclovir/pharmacology , Interleukin-10/genetics
3.
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
4.
Viruses ; 15(12)2023 12 15.
Article in English | MEDLINE | ID: mdl-38140679

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) establishes life-long latent infection and is linked to several human malignancies. Latency-associated nuclear antigen (LANA) is highly expressed during latency, and is responsible for the replication and maintenance of the viral genome. The expression of LANA is regulated at transcriptional/translational levels through multiple mechanisms, including the secondary structures in the mRNA sequence. LANA mRNA has multiple G-quadruplexes (G4s) that are bound by multiple proteins to stabilize/destabilize these secondary structures for regulating LANA. In this manuscript, we demonstrate the role of Nucleolin (NCL) in regulating LANA expression through its interaction with G-quadruplexes of LANA mRNA. This interaction reduced LANA's protein expression through the sequestration of mRNA into the nucleus, demonstrated by the colocalization of G4-carrying mRNA with NCL. Furthermore, the downregulation of NCL, by way of a short hairpin, showed an increase in LANA translation following an alteration in the levels of LANA mRNA in the cytoplasm. Overall, the data presented in this manuscript showed that G-quadruplexes-mediated translational control could be regulated by NCL, which can be exploited for controlling KSHV latency.


Subject(s)
G-Quadruplexes , Herpesvirus 8, Human , Humans , Herpesvirus 8, Human/physiology , Nucleolin , RNA, Messenger/genetics , RNA, Messenger/metabolism , Antigens, Viral/genetics , Virus Latency/genetics
5.
Int J Mol Sci ; 24(20)2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37894874

ABSTRACT

In eukaryotic organisms, genomic DNA associates with histone proteins to form nucleosomes. Nucleosomes provide a basis for genome compaction, epigenetic markup, and mediate interactions of nuclear proteins with their target DNA loci. A negatively charged (acidic) patch located on the H2A-H2B histone dimer is a characteristic feature of the nucleosomal surface. The acidic patch is a common site in the attachment of various chromatin proteins, including viral ones. Acidic patch-binding peptides present perspective compounds that can be used to modulate chromatin functioning by disrupting interactions of nucleosomes with natural proteins or alternatively targeting artificial moieties to the nucleosomes, which may be beneficial for the development of new therapeutics. In this work, we used several computational and experimental techniques to improve our understanding of how peptides may bind to the acidic patch and what are the consequences of their binding. Through extensive analysis of the PDB database, histone sequence analysis, and molecular dynamic simulations, we elucidated common binding patterns and key interactions that stabilize peptide-nucleosome complexes. Through MD simulations and FRET measurements, we characterized changes in nucleosome dynamics conferred by peptide binding. Using fluorescence polarization and gel electrophoresis, we evaluated the affinity and specificity of the LANA1-22 peptide to DNA and nucleosomes. Taken together, our study provides new insights into the different patterns of intermolecular interactions that can be employed by natural and designed peptides to bind to nucleosomes, and the effects of peptide binding on nucleosome dynamics and stability.


Subject(s)
Histones , Nucleosomes , Histones/metabolism , Fluorescence Resonance Energy Transfer , Chromatin , DNA/chemistry , Molecular Dynamics Simulation , Peptides/metabolism , Fluorescence Polarization
6.
Tumour Virus Res ; 15: 200259, 2023 06.
Article in English | MEDLINE | ID: mdl-36863485

ABSTRACT

Kaposi's Sarcoma (KS) is a heterogenous, multifocal vascular malignancy caused by the human herpesvirus 8 (HHV8), also known as Kaposi's Sarcoma-Associated Herpesvirus (KSHV). Here, we show that KS lesions express iNOS/NOS2 broadly throughout KS lesions, with enrichment in LANA positive spindle cells. The iNOS byproduct 3-nitrotyrosine is also enriched in LANA positive tumor cells and colocalizes with a fraction of LANA-nuclear bodies. We show that iNOS is highly expressed in the L1T3/mSLK tumor model of KS. iNOS expression correlated with KSHV lytic cycle gene expression, which was elevated in late-stage tumors (>4 weeks) but to a lesser degree in early stage (1 week) xenografts. Further, we show that L1T3/mSLK tumor growth is sensitive to an inhibitor of nitric oxide, L-NMMA. L-NMMA treatment reduced KSHV gene expression and perturbed cellular gene pathways relating to oxidative phosphorylation and mitochondrial dysfunction. These finding suggest that iNOS is expressed in KSHV infected endothelial-transformed tumor cells in KS, that iNOS expression depends on tumor microenvironment stress conditions, and that iNOS enzymatic activity contributes to KS tumor growth.


Subject(s)
Herpesvirus 8, Human , Sarcoma, Kaposi , Animals , Humans , Mice , Antigens, Viral/genetics , Herpesvirus 8, Human/genetics , omega-N-Methylarginine , Sarcoma, Kaposi/genetics , Tumor Microenvironment
7.
J Med Virol ; 95(1): e28255, 2023 01.
Article in English | MEDLINE | ID: mdl-36284455

ABSTRACT

Kaposi's sarcoma (KS) is the second most common tumor in people infected with human immunodeficiency virus worldwide, but its pathogenesis is still unclear. In this study, we discovered that the expression of GATA-binding protein 3 (GATA3) was lowly expressed in KS tissues and KSHV-infected cells, while microRNA-155 (miR-155) was highly expressed in KS serum and KSHV-infected cells. miR-155 promoted the proliferation, migration and invasion of KSHV infection by targeting GATA3. Further, The KSHV-encoded protein, the Latency associated nuclear antigen (LANA), promotes the proliferation, migration and invasion of KSHV-infected cells by regulating the miR-155/GATA3 axis. Regarding the molecular mechanism, c-Jun and c-Fos interact to form a complex. LANA upregulates the expression of c-Jun and c-Fos and enhances the formation of c-Jun/c-Fos complex. The complex binds to the -95∼-100 bp site of miR-155 promoter and transcriptionally activates miR-155. All in all, LANA enhances the c-Jun/c-Fos interaction, resulting in enhanced transcriptional regulation of miR-155 by the c-Jun/c-Fos complex, thereby downregulating GATA3 and promoting the proliferation, migration and invasion of KSHV-infected cells. The discovery of LANA/c-Jun/c-Fos/miR-155/GATA3 further refines the pathogenesis of KS, potentially opening a new avenue for developing effective drugs against KS.


Subject(s)
Herpesvirus 8, Human , MicroRNAs , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Cell Line , Antigens, Viral/metabolism , Antigens, Nuclear/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism
8.
Rev. peru. biol. (Impr.) ; 29(4)oct. 2022.
Article in English | LILACS-Express | LILACS | ID: biblio-1424296

ABSTRACT

The bee family Megachilidae consists of solitary species, some of which are important pollinators of cultivated plants. Although literature records indicate the existence of about 50 species of 10 genera of megachilid bees in Colombia, taxonomic studies are lacking and thus limited information is available on their identity as well as their distribution in the country. Herein, we provide new geographical records for the following ten species: Anthidium sanguinicaudum Schwarz, Chelostomoides otomita (Cresson), Hoplostelis bilineolata (Spinola), Megachile amparo Gonzalez, M. kalina Gonzalez et al., M. lorenziensis Mitchell, M. moderata Smith, M. simillima Smith, Pseudomegachile lanata (Fabricius), and Stelis costaricensis Friese. We report M. kalina for the first time for the country.


Las abejas de la familia Megachilidae son especies solitarias, algunas de ellas importantes polinizadores de cultivos. Aunque en la literatura se registran cerca de 50 especies de 10 géneros de abejas megachilidas en Colombia, faltan estudios taxonómicos y, por lo tanto, se dispone de información limitada sobre la identidad y la distribución de este grupo en el país. En este trabajo proporcionamos nuevos registros geográficos para 10 especies poco conocidas [Anthidium sanguinicaudum Schwarz, Chelostomoides otomita (Cresson), Hoplostelis bilineolata (Spinola), Megachile amparo Gonzalez, M. kalina Gonzalez et al., M. lorenziensis Mitchell, M. moderata Smith, M. simillima Smith, Pseudomegachile lanata (Fabricius), y Stelis costaricensis Friese]. Megachile kalina se registra por primera vez para Colombia.

9.
Cell Rep ; 40(7): 111234, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977517

ABSTRACT

Spermidine is essential for cellular growth and acts as a prerequisite of hypusination, a post-translational modification of eukaryotic initiation factor 5A (eIF5A), allowing the translation of polyproline-containing proteins. Here, we show that oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) increases spermidine synthesis and eIF5A hypusination to enhance expression of polyproline-containing latency-associated nuclear antigen (LANA) for viral episomal maintenance. KSHV upregulates intracellular spermidine levels by dysregulating polyamine metabolic pathways in three-dimensional (3D) culture and 2D de novo infection conditions. Increased intracellular spermidine leads to increased eIF5A hypusination, ultimately enhancing LANA expression. In contrast, inhibition of spermidine synthesis or eIF5A hypusination alleviates LANA expression, decreasing viral episomal maintenance and KSHV-infected cell proliferation in vitro and in vivo, which is reversed by spermidine supplement. This demonstrates that KSHV hijacks spermidine synthesis and eIF5A hypusination pathways to enhance LANA expression for viral episomal maintenance, suggesting polyamine metabolism and eIF5A hypusination as therapeutic targets for KSHV-induced tumorigenesis.


Subject(s)
Herpesvirus 8, Human , Spermidine , Antigens, Viral/metabolism , Cell Line , Herpesvirus 8, Human/physiology , Peptide Initiation Factors/metabolism , Protein Processing, Post-Translational , Spermidine/metabolism , Spermidine/pharmacology
10.
Protein Sci ; 31(6): e4339, 2022 06.
Article in English | MEDLINE | ID: mdl-35634775

ABSTRACT

Quantitative analysis of chromatin protein-nucleosome interactions is essential to understand regulation of genome-templated processes. However, current methods to measure nucleosome interactions are limited by low throughput, low signal-to-noise, and/or the requirement for specialized instrumentation. Here, we report a Lanthanide Chelate Excite Time-Resolved Fluorescence Resonance Energy Transfer (LANCE TR-FRET) assay to efficiently quantify chromatin protein-nucleosome interactions. The system makes use of commercially available reagents, offers robust signal-to-noise with minimal sample requirements, uses a conventional fluorescence microplate reader, and can be adapted for high-throughput workflows. We determined the nucleosome-binding affinities of several chromatin proteins and complexes, which are consistent with measurements obtained through orthogonal biophysical methods. We also developed a TR-FRET competition assay for high-resolution footprinting of chromatin protein-nucleosome interactions. Finally, we set up a TR-FRET competition assay using the LANA peptide to quantitate nucleosome acidic patch binding. We applied this assay to establish a proof-of-principle for regulation of nucleosome acidic patch binding by methylation of chromatin protein arginine anchors. Overall, our TR-FRET assays allow facile, high-throughput quantification of chromatin interactions and are poised to complement mechanistic chromatin biochemistry, structural biology, and drug discovery programs.


Subject(s)
Fluorescence Resonance Energy Transfer , Nucleosomes , Chromatin , Drug Discovery , Fluorescence Resonance Energy Transfer/methods
11.
Cell Rep ; 39(6): 110788, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545047

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) establishes a latent infection in the cell nucleus, but where KSHV episomal genomes are tethered and the mechanisms underlying KSHV lytic reactivation are unclear. Here, we study the nuclear microenvironment of KSHV episomes and show that the KSHV latency-lytic replication switch is regulated via viral long non-coding (lnc)RNA-CHD4 (chromodomain helicase DNA binding protein 4) interaction. KSHV episomes localize with CHD4 and ADNP proteins, components of the cellular ChAHP complex. The CHD4 and ADNP proteins occupy the 5'-region of the highly inducible lncRNAs and terminal repeats of the KSHV genome together with latency-associated nuclear antigen (LANA). Viral lncRNA binding competes with CHD4 DNA binding, and KSHV reactivation sequesters CHD4 from the KSHV genome, which is also accompanied by detachment of KSHV episomes from host chromosome docking sites. We propose a model in which robust KSHV lncRNA expression determines the latency-lytic decision by regulating LANA/CHD4 binding to KSHV episomes.


Subject(s)
Herpesvirus 8, Human , RNA, Long Noncoding , Sarcoma, Kaposi , Antigens, Viral/genetics , Antigens, Viral/metabolism , Chromosomes/metabolism , Herpesvirus 8, Human/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Plasmids , RNA, Long Noncoding/genetics , Tumor Microenvironment , Virus Latency/genetics
12.
mBio ; 13(2): e0173321, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35343785

ABSTRACT

The past decade illuminated the H2A-H2B acidic patch as a cornerstone for both nucleosome recognition and chromatin structure regulation. Higher-order folding of chromatin arrays is mediated by interactions of histone H4 tail with an adjacent nucleosome acidic patch. Dynamic chromatin folding ensures a proper regulation of nuclear functions fundamental to cellular homeostasis. Many cellular factors have been shown to act on chromatin by tethering nucleosomes via an arginine anchor binding to the acidic patch. This tethering mechanism has also been described for several viral proteins. In this minireview, we will discuss the structural basis for acidic patch engagement by viral proteins and the implications during respective viral infections. We will also discuss a model in which acidic patch occupancy by these non-self viral proteins alters the local chromatin state by preventing H4 tail-mediated higher-order chromatin folding.


Subject(s)
Nucleosomes , Viral Proteins , Chromatin , Histones/metabolism , Viral Proteins/metabolism
13.
FEBS J ; 289(24): 7631-7669, 2022 12.
Article in English | MEDLINE | ID: mdl-34536980

ABSTRACT

Epstein-Barr virus (EBV; human herpesvirus 4; HHV-4) and Kaposi sarcoma-associated herpesvirus (KSHV; human herpesvirus 8; HHV-8) are human gammaherpesviruses that have oncogenic properties. EBV is a lymphocryptovirus, whereas HHV-8/KSHV is a rhadinovirus. As lymphotropic viruses, EBV and KSHV are associated with several lymphoproliferative diseases or plasmacytic/plasmablastic neoplasms. Interestingly, these viruses can also infect epithelial cells causing carcinomas and, in the case of KSHV, endothelial cells, causing sarcoma. EBV is associated with Burkitt lymphoma, classic Hodgkin lymphoma, nasopharyngeal carcinoma, plasmablastic lymphoma, lymphomatoid granulomatosis, leiomyosarcoma, and subsets of diffuse large B-cell lymphoma, post-transplant lymphoproliferative disorder, and gastric carcinoma. KSHV is implicated in Kaposi sarcoma, primary effusion lymphoma, multicentric Castleman disease, and KSHV-positive diffuse large B-cell lymphoma. Pathogenesis by these two herpesviruses is intrinsically linked to viral proteins expressed during the lytic and latent lifecycles. This comprehensive review intends to provide an overview of the EBV and KSHV viral cycles, viral proteins that contribute to oncogenesis, and the current understanding of the pathogenesis and clinicopathology of their related neoplastic entities.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 8, Human , Lymphoma, Large B-Cell, Diffuse , Sarcoma, Kaposi , Humans , Epstein-Barr Virus Infections/complications , Endothelial Cells/pathology , Herpesvirus 4, Human/genetics , Sarcoma, Kaposi/pathology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
14.
Cancers (Basel) ; 13(24)2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34944828

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV), also called human herpesvirus 8 (HHV-8), is an oncogenic virus belonging to the Herpesviridae family. The viral particle is composed of a double-stranded DNA harboring 90 open reading frames, incorporated in an icosahedral capsid and enveloped. The viral cycle is divided in the following two states: a short lytic phase, and a latency phase that leads to a persistent infection in target cells and the expression of a small number of genes, including LANA-1, v-FLIP and v-cyclin. The seroprevalence and risk factors of infection differ around the world, and saliva seems to play a major role in viral transmission. KSHV is found in all epidemiological forms of Kaposi's sarcoma including classic, endemic, iatrogenic, epidemic and non-epidemic forms. In a Kaposi's sarcoma lesion, KSHV is mainly in a latent state; however, a small proportion of viral particles (<5%) are in a replicative state and are reported to be potentially involved in the proliferation of neighboring cells, suggesting they have crucial roles in the process of tumorigenesis. KSHV encodes oncogenic proteins (LANA-1, v-FLIP, v-cyclin, v-GPCR, v-IL6, v-CCL, v-MIP, v-IRF, etc.) that can modulate cellular pathways in order to induce the characteristics found in all cancer, including the inhibition of apoptosis, cells' proliferation stimulation, angiogenesis, inflammation and immune escape, and, therefore, are involved in the development of Kaposi's sarcoma.

15.
mSystems ; 6(6): e0110921, 2021 Dec 21.
Article in English | MEDLINE | ID: mdl-34726485

ABSTRACT

Hypoxia signaling is a key regulator in the development and progression of many types of human malignancies, including viral cancers. The latency-associated nuclear antigen (LANA), encoded by Kaposi's sarcoma-associated herpesvirus (KSHV) during latency, is a multifunctional protein that plays an essential role in viral episome maintenance and lytic gene silencing for inducing tumorigenesis. Although our previous studies have shown that LANA contains a SUMO-interacting motif (LANASIM), and hypoxia reduces SUMOylated KAP1 association with LANASIM, the physiological proteomic network of LANASIM-associated cellular proteins in response to hypoxia is still unclear. In this study, we individually established cell lines stably expressing wild-type LANA (LANAWT) and its SIM-deleted mutant (LANAdSIM) and treated them with or without hypoxia, followed by coimmunoprecipitation and mass spectrometry analysis to systemically identify the hypoxia-responsive profile of LANASIM-associated cellular proteins. We found that in hypoxia, the number of cellular proteins associated with LANAWT instead of LANAdSIM was dramatically increased. Functional network analysis revealed that two major pathways, which included cytoskeleton organization and DNA/RNA binding and processing pathways, were significantly enriched for 28 LANASIM-associated proteins in response to hypoxia. HNRNPU was one of the proteins consistently identified that interacted with LANASIM in different proteomic screening systems and responded to hypoxia. This study provides a proteomic profile of LANASIM-associated proteins in hypoxia and facilitates our understanding of the role of the collaboration between viral infection and the hypoxia response in inducing viral persistence and tumorigenesis. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) has been reported to be involved in the regulation of host proteins in response to hypoxic stress. LANA, one of the key latent proteins, contains a SUMO-interacting motif (LANASIM) and reduces the association with SUMOylated KAP1 upon hypoxic treatment. However, the physiological systematic network of LANASIM-associated cellular proteins in hypoxia is still unclear. Here, we revealed two major pathways, which included cytoskeleton organization and DNA/RNA binding and processing pathways, that were significantly enriched for 28 LANASIM-associated proteins in hypoxia. This discovery not only provides a proteomic profile of LANASIM-associated proteins in hypoxia but also facilitates our understanding of the collaboration between viral infection and hypoxic stress in inducing viral persistence and tumorigenesis.

16.
Int J Mol Sci ; 22(21)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34769420

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is a cancer-related virus which engages in two forms of infection: latent and lytic. Latent infection allows the virus to establish long-term persistent infection, whereas the lytic cycle is needed for the maintenance of the viral reservoir and for virus spread. By using recombinant KSHV viruses encoding mNeonGreen and mCherry fluorescent proteins, we show that various cell types that are latently-infected with KSHV can be superinfected, and that the new incoming viruses establish latent infection. Moreover, we show that latency establishment is enhanced in superinfected cells compared to primary infected ones. Further analysis revealed that cells that ectopically express the major latency protein of KSHV, LANA-1, prior to and during infection exhibit enhanced establishment of latency, but not cells expressing LANA-1 fragments. This observation supports the notion that the expression level of LANA-1 following infection determines the efficiency of latency establishment and avoids loss of viral genomes. These findings imply that a host can be infected with more than a single viral genome and that superinfection may support the maintenance of long-term latency.


Subject(s)
Antigens, Viral/metabolism , Herpesvirus 8, Human/physiology , Nuclear Proteins/metabolism , Sarcoma, Kaposi/virology , Superinfection/virology , Cell Line , Genome, Viral , Humans , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/pathology , Superinfection/genetics , Superinfection/pathology , Virus Latency
17.
Viruses ; 13(9)2021 09 09.
Article in English | MEDLINE | ID: mdl-34578378

ABSTRACT

Kaposi-sarcoma-associated herpesvirus (KSHV) or human herpesvirus 8 (HHV-8) is the causative agent of several malignancies, including Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD). Active KSHV replication has also been associated with a pathological condition called KSHV inflammatory cytokine syndrome (KICS), and KSHV may play a role in rare cases of post-transplant polyclonal lymphoproliferative disorders. Several commonly used herpesviral DNA polymerase inhibitors are active against KSHV in tissue culture. Unfortunately, they are not always efficacious against KSHV-induced diseases. To improve the outcome for the patients, new therapeutics need to be developed, including treatment strategies that target either viral proteins or cellular pathways involved in tumor growth and/or supporting the viral life cycle. In this review, we summarize the most commonly established treatments against KSHV-related diseases and review recent developments and promising new compounds that are currently under investigation or on the way to clinical use.


Subject(s)
Herpesviridae Infections/drug therapy , Herpesvirus 8, Human/drug effects , Sarcoma, Kaposi/drug therapy , Virus Replication/genetics , Animals , CRISPR-Cas Systems , Castleman Disease/drug therapy , Clinical Trials as Topic , DNA-Directed DNA Polymerase , Exodeoxyribonucleases/antagonists & inhibitors , Gene Expression Regulation, Viral , Herpesviridae Infections/classification , Herpesvirus 8, Human/genetics , Humans , Lymphoma, Primary Effusion/drug therapy , Mice , Sarcoma, Kaposi/virology , Viral Proteins/antagonists & inhibitors , Virus Latency/genetics , Virus Replication/drug effects
18.
Future Med Chem ; 13(13): 1141-1151, 2021 07.
Article in English | MEDLINE | ID: mdl-34036806

ABSTRACT

The Kaposi sarcoma-associated herpesvirus (KSHV) is an oncogenic human herpesviruses that is responsible for cancer, especially in immunosuppressed people, such as patients with AIDS. So far, there are no KSHV-specifc antiviral agents available. In this review, we provide an overview on one particular target-centered approach toward novel anti-KSHV drugs focusing on interfering with the molecular functions of the latency-associated nuclear antigen (LANA). This review focuses on attempts to interfere with the LANA-DNA interaction mediated by the C-terminal domain. We describe the drug discovery approaches chosen for this endeavor as well as molecular structures that were identified in this innovative concept toward novel and KSHV-specific antiherpesviral agents.


Subject(s)
Antiviral Agents/pharmacology , DNA, Viral/drug effects , Herpesvirus 8, Human/drug effects , Nuclear Proteins/antagonists & inhibitors , Antigens, Viral , Antiviral Agents/chemistry , Humans , Microbial Sensitivity Tests
19.
J Med Virol ; 93(8): 5065-5075, 2021 08.
Article in English | MEDLINE | ID: mdl-33942339

ABSTRACT

Viral infection has been implicated in the pathogenesis of a plethora of human diseases. Although antiviral therapies effectively confront the viral spread and infection, how to completely eradicate the viral genome from infected cells remains a challenge. In this study, we demonstrated the reversible switching of primary cells between normal and malignant states by an oncogenic virus Kaposi's sarcoma-associated herpesvirus (KSHV) and CRISPR/Cas9-mediated targeting of a major viral latent protein. Primary cells can be transformed into malignant status by infection of KSHV, while elimination of the KSHV genome from latent KSHV-infected cells reverses KSHV-transformed primary cells back to a "normal state" by CRISPR/Cas-mediated knockout of viral major latent gene LANA. As a proof of concept, we demonstrated efficient elimination of KSHV episome in KSHV-associated primary effusion lymphoma cells resulting in the induction of apoptosis by liposome-encapsulated CRISPR/Cas9 ribonucleoprotein complexes (Lipo/Cas9-LANAsgRNA). Our work illustrates CRISPR/Cas as a promising technology for eliminating oncogenic viruses from persistently infected cells by taking advantage of the genetic differences between viral and cellular genomes. Compared to traditional antiviral therapy, our study offer an approach for antagonizing human oncogenic virus-related cancers by directly targeting as well as clearing viral genomes.


Subject(s)
Antigens, Viral/genetics , CRISPR-Cas Systems , Cell Transformation, Neoplastic/genetics , Herpesvirus 8, Human/genetics , Nuclear Proteins/genetics , Oncogenic Viruses/genetics , Animals , Antigens, Viral/metabolism , Apoptosis , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Cell Cycle , Cell Proliferation , Gene Knockout Techniques , Genome, Viral/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, Primary Effusion/pathology , Mesenchymal Stem Cells , Nuclear Proteins/metabolism , Oncogenic Viruses/pathogenicity , RNA, Guide, Kinetoplastida/genetics , Rats , Virus Latency/genetics
20.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: mdl-33789996

ABSTRACT

The hypoxic microenvironment and metabolic reprogramming are two major contributors to the phenotype of oncogenic virus-infected cells. Infection by Kaposi's sarcoma-associated herpesvirus (KSHV) stabilizes hypoxia-inducible factor 1α (HIF1α) and reprograms cellular metabolism. We investigated the comparative transcriptional regulation of all major genes involved in fatty acid and amino acid metabolism in KSHV-positive and -negative cells grown under normoxic or hypoxic conditions. We show a distinct regulation of genes involved in both fatty acid and amino acid metabolism in KSHV-positive cells grown in either normoxic or hypoxic conditions, with a particular focus on genes involved in the acetyl coenzyme A (acetyl-CoA) pathway. The fatty acid binding protein (FABP) family of genes, specifically FABP1, FABP4, and FABP7, was also observed to be synergistically upregulated in hypoxia by KSHV. This pattern of FABP gene expression was also seen in naturally infected KSHV BC3 or BCBL1 cells when compared to KSHV-negative DG75 or BL41 cells. Two KSHV-encoded antigens, which positively regulate HIF1α, the viral G-protein coupled receptor (vGPCR), and the latency-associated nuclear antigen (LANA) were shown to drive upregulation of the FABP gene transcripts. Suppression of FABPs by RNA interference resulted in an adverse effect on hypoxia-dependent viral reactivation. Overall, this study provides new evidence, which supports a rationale for the inhibition of FABPs in KSHV-positive cells as potential strategies, for the development of therapeutic approaches targeting KSHV-associated malignancies.IMPORTANCE Hypoxia is a detrimental stress to eukaryotes and inhibits several cellular processes, such as DNA replication, transcription, translation, and metabolism. Interestingly, the genome of Kaposi's sarcoma-associated herpesvirus (KSHV) is known to undergo productive replication in hypoxia. We investigated the comparative transcriptional regulation of all major genes involved in fatty acid and amino acid metabolism in KSHV-positive and -negative cells grown under normoxic or hypoxic conditions. Several metabolic pathways were observed differentially regulated by KSHV in hypoxia, specifically, the fatty acid binding protein (FABP) family genes (FABP1, FABP4, and FABP7). KSHV-encoded antigens, vGPCR and LANA, were shown to drive upregulation of the FABP transcripts. Suppression of FABPs by RNA interference resulted in an adverse effect on hypoxia-dependent viral reactivation. Overall, this study provides new evidence, which supports a rationale for the inhibition of FABPs in KSHV-positive cells as potential strategies, for the development of therapeutic approaches targeting KSHV-associated malignancies.


Subject(s)
Cell Hypoxia , Fatty Acid-Binding Protein 7/genetics , Fatty Acid-Binding Proteins/genetics , Herpesvirus 8, Human/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Tumor Suppressor Proteins/genetics , Amino Acids/metabolism , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cell Line, Tumor , Fatty Acid-Binding Protein 7/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Tumor Suppressor Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Activation
SELECTION OF CITATIONS
SEARCH DETAIL