Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters








Publication year range
1.
Front Immunol ; 15: 1329846, 2024.
Article in English | MEDLINE | ID: mdl-38529279

ABSTRACT

Understanding adaptive immunity against SARS-CoV-2 is a major requisite for the development of effective vaccines and treatments for COVID-19. CD4+ T cells play an integral role in this process primarily by generating antiviral cytokines and providing help to antibody-producing B cells. To empower detailed studies of SARS-CoV-2-specific CD4+ T cell responses in mouse models, we comprehensively mapped I-Ab-restricted epitopes for the spike and nucleocapsid proteins of the BA.1 variant of concern via IFNγ ELISpot assay. This was followed by the generation of corresponding peptide:MHCII tetramer reagents to directly stain epitope-specific T cells. Using this rigorous validation strategy, we identified 6 immunogenic epitopes in spike and 3 in nucleocapsid, all of which are conserved in the ancestral Wuhan strain. We also validated a previously identified epitope from Wuhan that is absent in BA.1. These epitopes and tetramers will be invaluable tools for SARS-CoV-2 antigen-specific CD4+ T cell studies in mice.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Mice , CD4-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , Nucleocapsid/chemistry , Peptides/chemistry , SARS-CoV-2/chemistry , Histocompatibility Antigens Class II/chemistry , Spike Glycoprotein, Coronavirus/chemistry
2.
J Med Virol ; 95(3): e28676, 2023 03.
Article in English | MEDLINE | ID: mdl-36929740

ABSTRACT

Reactive oxygen species (ROS) are a group of a highly short-lived molecules that control diverse behaviors of cells. Normal cells maintain ROS balance to ensure their functions. Because of oncogenic stress, cancer cells often have excessive ROS, also known as oxidative stress, which are often counteracted by enhanced antioxidant systems to maintain redox homeostasis. Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus associated with Kaposi's sarcoma (KS), which manifests hyper inflammation and oxidative stress as the hallmarks. We have previously shown that excessive ROS can disrupt KSHV latency by inducing viral lytic replication, leading to cell death. Paradoxically, most KS tumor cells are latently infected by KSHV in a highly inflammatory and oxidative stress tumor microenvironment, which is in part due to the activation of alternative complement and TLR4 pathways, indicating the existence of an enhanced antioxidant defense system in KS tumor cells. In this study, we show that KSHV upregulates antioxidant genes, including SOD2 and CAT by hijacking the forkhead box protein O1 (FoxO1), to maintain intracellular ROS level. Moreover, the fine-tuned balance of ROS level in KSHV-transformed cells is essential for cell survival. Consequently, KSHV-transformed cells are extremely sensitive to exogenous ROS insult such as treatment with a low level of hydrogen peroxide (H2 O2 ). Either chemical inhibition or knockdown of FoxO1 by short interfering RNAs decreases the expression of antioxidant genes and subsequently increases the intracellular ROS level in KSHV-transformed cells, resulting in the inhibition of cell proliferation and colony formation in soft agar. Mechanistically, KSHV-encoded microRNAs and vFLIP upregulate FoxO1 by activating the NF-κB pathway. These results reveal a novel mechanism by which an oncogenic virus counteracts oxidative stress by upregulating FoxO1, which is essential for KSHV-induced cell proliferation and cellular transformation. Therefore, FoxO1 might be a potential therapeutic target for KSHV-related malignancies.


Subject(s)
Herpesvirus 8, Human , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Reactive Oxygen Species , Antioxidants/metabolism , Oxidative Stress , Cell Proliferation , Tumor Microenvironment , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism
3.
Cell Oncol (Dordr) ; 45(5): 967-989, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35964258

ABSTRACT

BACKGROUND: Primary effusion lymphoma (PEL) is an aggressive form of non-Hodgkin lymphoma of B cells caused by Kaposi's Sarcoma-associated Herpes Virus (KSHV). KSHV encoded latent and lytic antigens promote oncogenic transformation and evade apoptosis through the modulation of various host cellular signaling pathways. Nm23-H1 is a known metastatic suppressor whose expression inversely correlates with the metastatic potential of different cancers. Here, we set out to assess the role of Nm23-H1 in PEL development. METHODS: Flow cytometry and real-time PCR assays were performed for Nm23-H1 expression analysis. Induction of apoptosis was assessed using Western blotting and flow cytometry-based assays in Nm23-H1 overexpressing cells. Co-immunoprecipitation assays, confocal microscopy and imaging flow cytometry were performed to determine Nm23-H1 and vFLIP K13 protein-protein interaction. A PEL cell-induced xenograft model was established in non-obese diabetic/severely combined immunodeficient (NOD/SCID) mice to validate the effect of Nm23-H1 overexpression. RESULTS: We found that Nm23-H1 expression was significantly downregulated both at transcriptional and protein levels in PEL cell lines and that its overexpression triggered mitochondrial-mediated caspase-dependent apoptosis. We revealed Nm23-H1 interacts with the latent protein vFLIP K13 and that Nm23-H1 overexpression leads to inhibition of vFLIP K13 driven nuclear factor-kappa B (NF-κB) signaling with concurrent inhibition of autocrine and paracrine growth factors and downregulation of latent KSHV antigens without induction of active lytic reactivation. We also confirmed the effects of Nm23-H1 overexpression in a PEL cell-induced xenograft model in NOD/SCID mice. CONCLUSION: Downregulation of Nm23-H1 expression in KSHV-infected PEL cells and its overexpression trigger apoptosis by impairing vFLIP K13-driven NF-κB signaling, suggesting therapeutic implications of Nm23-H1 for primary effusion lymphomas.


Subject(s)
Herpesvirus 8, Human , Lymphoma, Primary Effusion , Sarcoma, Kaposi , Animals , Humans , Mice , Apoptosis , Herpesvirus 8, Human/metabolism , Lymphoma, Primary Effusion/metabolism , Mice, Inbred NOD , Mice, SCID , NF-kappa B/metabolism , Oncogene Proteins/metabolism , Sarcoma, Kaposi/metabolism , Viral Proteins/metabolism
4.
Antiviral Res ; 204: 105370, 2022 08.
Article in English | MEDLINE | ID: mdl-35772601

ABSTRACT

Next-generation COVID-19 vaccines are critical due to the ongoing evolution of SARS-CoV-2 virus and rapid waning duration of the neutralizing antibody response against current vaccines. The mRNA vaccines mRNA-1273 and BNT162b2 were developed using linear transcripts encoding the prefusion-stabilized trimers (S-2P) of the wildtype spike, which have shown a reduced neutralizing activity against the variants of concern B.1.617.2 and B.1.1.529. Recently, a new version of spike trimer, termed VFLIP (five (V) prolines, Flexibly-Linked, Inter-Protomer disulfide) was developed. Based on the original amino acid sequence of the wildtype spike, VFLIP was genetically engineered by using five proline substitutions, a flexible cleavage site amino acid linker, and an inter-protomer disulfide bond. It has been suggested to possess native-like glycosylation, and greater pre-fusion trimeric stability as opposed to S-2P. Here, we report that the spike protein VFLIP-X, containing six rationally substituted amino acids to reflect emerging variants (K417N, L452R, T478K, E484K, N501Y and D614G), offers a promising candidate for a next-generation SARS-CoV-2 vaccine. Mice immunized by a circular mRNA (circRNA) vaccine prototype producing VFLIP-X had detectable neutralizing antibody titers for up to 7 weeks post-boost against SARS-CoV-2 variants of concern (VOCs) and variants of interest (VOIs). In addition, a balance in TH1 and TH2 responses was achieved by immunization with VFLIP-X. Our results indicate that the VFLIP-X delivered by circRNA induces humoral and cellular immune responses, as well as broad neutralizing activity against SARS-CoV-2 variants.


Subject(s)
COVID-19 Vaccines , COVID-19 , RNA, Circular , SARS-CoV-2 , mRNA Vaccines , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines/genetics , Disulfides , Mice , Proline , Protein Subunits , RNA, Circular/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , mRNA Vaccines/genetics
5.
J Biol Chem ; 298(6): 102012, 2022 06.
Article in English | MEDLINE | ID: mdl-35525271

ABSTRACT

Constitutive activation of the canonical NF-κB signaling pathway is a major factor in Kaposi's sarcoma-associated herpes virus pathogenesis where it is essential for the survival of primary effusion lymphoma. Central to this process is persistent upregulation of the inhibitor of κB kinase (IKK) complex by the virally encoded oncoprotein vFLIP. Although the physical interaction between vFLIP and the IKK kinase regulatory component essential for persistent activation, IKKγ, has been well characterized, it remains unclear how the kinase subunits are rendered active mechanistically. Using a combination of cell-based assays, biophysical techniques, and structural biology, we demonstrate here that vFLIP alone is sufficient to activate the IKK kinase complex. Furthermore, we identify weakly stabilized, high molecular weight vFLIP-IKKγ assemblies that are key to the activation process. Taken together, our results are the first to reveal that vFLIP-induced NF-κB activation pivots on the formation of structurally specific vFLIP-IKKγ multimers which have an important role in rendering the kinase subunits active through a process of autophosphorylation. This mechanism of NF-κB activation is in contrast to those utilized by endogenous cytokines and cellular FLIP homologues.


Subject(s)
Herpesvirus 8, Human , Sarcoma, Kaposi , Enzyme Activation/genetics , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , I-kappa B Kinase/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oncogene Proteins/metabolism , Sarcoma, Kaposi/enzymology , Sarcoma, Kaposi/virology , Viral Proteins/metabolism
6.
J Med Virol ; 94(6): 2766-2775, 2022 06.
Article in English | MEDLINE | ID: mdl-35149992

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) causes life-long latent infection and malignancies, including KS commonly found in AIDS patients. Lytic replication can be induced to kill tumor cells harboring latent KSHV, through viral cytopathic effects and the subsequent antiviral immune responses. Viral FLICE-inhibitory protein (vFLIP), encoded by KSHV ORF K13, inhibits KSHV lytic reactivation, implying that the competing endogenous RNA (ceRNA) networks regulated by vFLIP can be modulated to induce the lytic reactivation of latent KSHV, a promising strategy for KSHV-associated malignancies. Here, we performed whole-transcriptome sequencing to reveal the global landscape of noncoding RNAs and messenger RNAs (mRNAs) in iSLK-RGB-BAC16 cells and iSLK-RGB-K13 mutant cells. It showed that vFLIP regulated 227 differentially expressed (DE) long non-coding RNAs (lncRNAs), 57 DE circular RNAs (circRNAs), 20 DE microRNAs (miRNAs), and 1371 DE mRNAs. Enrichment analysis verified that riboflavin metabolism was simultaneously enriched in DE genes related to miRNAs, lncRNAs, and circRNAs. The upregulated hsa-miR-378i and hsa-miR-3654, and downregulated miR-4467, miR-3163, miR-4451, and miR-4257 were significantly enriched in the ceRNA complex network, which contained 9 upregulated and 7 downregulated circRNAs, 5 upregulated and 85 downregulated lncRNAs, 5 upregulated and 35 downregulated mRNAs. Finally, we constructed and validated two vFLIP-regulated ceRNA networks: circRNA hsa_circ_0070049/hsa-miR-378i/SPEG/FOXQ1 and lncRNA AL031123.1/hsa-miR-378i/SPEG/FOXQ1. Taken together, the two ceRNA networks may mediate KSHV reactivation. These novel findings refreshed the present understanding of ceRNA network in KSHV lytic induction and provided potential therapeutic targets for KSHV-associated malignancies.


Subject(s)
Herpesvirus 8, Human , MicroRNAs , Neoplasms , RNA, Long Noncoding , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Forkhead Transcription Factors , Herpesvirus 8, Human/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Viral Proteins/metabolism
7.
Viruses ; 14(2)2022 02 11.
Article in English | MEDLINE | ID: mdl-35215970

ABSTRACT

During viral evolution and adaptation, many viruses have utilized host cellular factors and machinery as their partners. HBx, as a multifunctional viral protein encoded by the hepatitis B virus (HBV), promotes HBV replication and greatly contributes to the development of HBV-associated hepatocellular carcinoma (HCC). HBx interacts with several host factors in order to regulate HBV replication and evolve carcinogenesis. The cellular FADD-like IL-1ß-converting enzyme (FLICE)-like inhibitory protein (c-FLIP) is a major factor that functions in a variety of cellular pathways and specifically in apoptosis. It has been shown that the interaction between HBx and c-FLIP determines HBV fate. In this review, we provide a comprehensive and detailed overview of the interplay between c-FLIP and HBV in various environmental circumstances. We describe strategies adapted by HBV to establish its chronic infection. We also summarize the conventional roles of c-FLIP and highlight the functional outcome of the interaction between c-FLIP and HBV or other viruses in viral replication and the innate immune system.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Hepatitis B virus/physiology , Host-Pathogen Interactions , Immune System/metabolism , Virus Replication , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Carcinoma, Hepatocellular/virology , Hepatocytes/virology , Humans , Liver Neoplasms/virology , Viral Regulatory and Accessory Proteins
8.
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
9.
Microb Pathog ; 144: 104170, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32224211

ABSTRACT

Some viruses encode inhibitory factors of apoptosis during infection to prolong cell viability and then to achieve a higher production of viral progeny or facilitate persistent infections. There is evidence that some gammaherpesviruses, including BoHV-4, carry genes that can both inhibit or induce apoptosis. BoHV-4 possesses two genes (ORF16 and ORF71) that code for proteins with anti-apoptotic functions, such as v-Bcl2 and v-Flip, respectively. Thus, it is relevant to study BoHV-4 in relation to the modulation of apoptosis in infected cells as a strategy for persistence in the host. The objective of this work was to analyze whether variations in v-Flip and v- Bcl2 of six phylogenetically divergent Argentinean isolates of BoHV-4 can influence the capacity of these strains to induce apoptosis in cell cultures. In this study, variations were mainly detected in the v-Flip gene and protein of the BoHV-4 strains belonging to genotype 3. Thus, it is possible to infer that sequence variations could be associated with some BoHV-4 genotype. Induction of apoptosis was not a significant event for any of the genetically distinct local isolates of BoHV-4 and there was not an evident relationship between the variability of both genes with the apoptotic effect of the phylogenetically distinct strains.


Subject(s)
Apoptosis/genetics , Herpesvirus 4, Bovine/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Viral Proteins/genetics , Amino Acid Sequence , Animals , Argentina , Base Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cattle , Cattle Diseases/virology , Cell Line, Tumor , Genotype , HeLa Cells , Herpesviridae Infections/veterinary , Herpesvirus 4, Bovine/isolation & purification , Humans , Sequence Alignment
10.
BMC Genomics ; 20(Suppl 3): 293, 2019 May 08.
Article in English | MEDLINE | ID: mdl-31815628

ABSTRACT

BACKGROUND: Structural homology modeling supported by bioinformatics analysis plays a key role in uncovering new molecular interactions within gene regulatory networks. Here, we have applied this powerful approach to analyze the molecular interactions orchestrating death receptor signaling networks. In particular, we focused on the molecular mechanisms of CD95-mediated NF-κB activation and the role of c-FLIP/NEMO interaction in the induction of this pathway. RESULTS: To this end, we have created the homology model of the c-FLIP/NEMO complex using the reported structure of the v-FLIP/NEMO complex, and rationally designed peptides targeting this complex. The designed peptides were based on the NEMO structure. Strikingly, the experimental in vitro validation demonstrated that the best inhibitory effects on CD95-mediated NF-κB activation are exhibited by the NEMO-derived peptides with the substitution D242Y of NEMO. Furthermore, we have assumed that the c-FLIP/NEMO complex is recruited to the DED filaments formed upon CD95 activation and validated this assumption in silico. Further insight into the function of c-FLIP/NEMO complex was provided by the analysis of evolutionary conservation of interacting regions which demonstrated that this interaction is common in distinct mammalian species. CONCLUSIONS: Taken together, using a combination of bioinformatics and experimental approaches we obtained new insights into CD95-mediated NF-κB activation, providing manifold possibilities for targeting the death receptor network.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , I-kappa B Kinase/metabolism , Molecular Probes , NF-kappa B/metabolism , fas Receptor/metabolism , Amino Acid Sequence , Computational Biology , Humans , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Sequence Alignment , Signal Transduction
11.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30842330

ABSTRACT

MC159 is a viral FLIP (FLICE inhibitory protein) encoded by the molluscum contagiosum virus (MCV) enabling MCV to evade antiviral immunity and to establish persistent infections in humans. Here, we show that MC159 contains a functional SH3 binding motif, which mediates avid and selective binding to SH3BP4, a signaling protein known to regulate endocytic trafficking and suppress cellular autophagy. The capacity to bind SH3BP4 was dispensable for regulation of NF-κB-mediated transcription and suppression of proapoptotic caspase activation but contributed to inhibition of amino acid starvation-induced autophagy by MC159. These results provide new insights into the cellular functions of MC159 and reveal SH3BP4 as a novel host cell factor targeted by a viral immune evasion protein.IMPORTANCE After the eradication of smallpox, molluscum contagiosum virus (MCV) is the only poxvirus restricted to infecting humans. MCV infection is common and causes benign skin lesions that usually resolve spontaneously but may persist for years and grow large, especially in immunocompromised individuals. While not life threatening, MCV infections pose a significant global health burden. No vaccine or specific anti-MCV therapy is available. MCV encodes several proteins that enable it to evade antiviral immunity, a notable example of which is the MC159 protein. In this study, we describe a novel mechanism of action for MC159 involving hijacking of a host cell protein called SH3BP4 to suppress autophagy, a cellular recycling mechanism important for antiviral immunity. This study contributes to our understanding of the host cell interactions of MCV and the molecular function of MC159.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Molluscum contagiosum virus/metabolism , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/physiology , Apoptosis/drug effects , Autophagy/physiology , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/drug effects , Humans , Immune Evasion/drug effects , Immune Evasion/physiology , MCF-7 Cells , Molluscum Contagiosum/virology , Molluscum contagiosum virus/pathogenicity , NF-kappa B/metabolism , Protein Binding , Protein Processing, Post-Translational/drug effects , Signal Transduction , Viral Proteins/physiology , src Homology Domains/physiology
12.
Virology ; 522: 106-121, 2018 09.
Article in English | MEDLINE | ID: mdl-30029010

ABSTRACT

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) vFLIP, a latent gene of KSHV, was first identified as a FLICE-inhibitory protein (FLIP) protecting cells from apoptosis. The vFLIP protein has been shown to activate the NF-κB signaling involved in spindle morphology formation both in HUVECs infected with KSHV and Kaposi's sarcoma (KS) itself. In this study, we independently established stably vFLIP-expressing cells and showed that they exhibited upregulated NF-κB family protein expression independent of the ability of IKKs to bind vFLIP. Further, vFLIP induced upregulation of IKKε, phosphorylation of RelA at Ser468 (p-RelA S468) and nuclear localization of Re1A concomitant with spindle morphology formation, and these effects were reversed by knockdown of IKKε and treatment with Bay-11. Overexpression of IKKε alone also showed spindle morphology formation with p-RelA S468. In conclusion, the spindle cell morphology in KS should be induced by RelA activation (p-RelA S468) by IKKε upregulation in vFLIP-expressing EA hy926 cells.


Subject(s)
Epithelial Cells/cytology , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , I-kappa B Kinase/metabolism , Transcription Factor RelA/metabolism , Viral Proteins/metabolism , Cell Line , Epithelial Cells/virology , Humans , Up-Regulation
13.
Autophagy ; 14(8): 1359-1375, 2018.
Article in English | MEDLINE | ID: mdl-29962265

ABSTRACT

Although antiretroviral therapy is highly effective in suppressing human immunodeficiency virus type-1 (HIV) replication, treatment has failed to eliminate viral reservoirs and discontinuation of treatment results in viral reactivation. Here, we demonstrate that peptides Tat-vFLIP-α2 and Tat-Beclin 1/BECN1 which have been shown to induce a Na+/K+-ATPase- and a macroautophagy/autophagy-dependent form of cell death, autosis, can preferentially kill HIV-infected macrophages while preventing virological rebound. To improve bioavailability and drug delivery, Tat-vFLIP-α2 was encapsulated into biodegradable PLGA (poly lactic-co-glycolic acid)-lipid-PEG (polyethylene glycol) nanoparticles for long-lasting intracellular delivery. After a single dose of NP-vFLIP-α2, HIV-infected macrophages were preferentially killed in a dose-dependent manner compared to uninfected or untreated HIV-infected cells with complete inhibition of HIV infection at 10 µM of peptide. HIV-infected macrophages treated with NP-vFLIP-α2 exhibited increased markers of autophagy including LC3B lipidation, SQSTM1/p62 degradation and Na+/K+-ATPase expression compared to untreated uninfected or infected cells. Moreover, the increased cell death observed in HIV-infected cells was not altered by treatment with bafilomycin A1 (BAF) or the caspase inhibitor Z-VAD-FMK, but could be reversed following treatment with the Na+/K+-ATPase inhibitor, digoxin, or knockdown of ATG5 or ATG7. NP-vFLIP-α2 induced preferential killing was also detected in HIV-infected macrophages under antiretroviral suppression without inducing viral reactivation. Additionally, we found that Na+/K+-ATPase was upregulated in HIV-infected cells, which enhanced NP-vFLIP-α2 induced cell death. These findings provide a novel strategy to eradicate HIV-infected macrophages by selectively killing infected cells through the induction of Na+/K+-ATPase dependent autophagy, while preventing reactivation of virus and new infection of uninfected bystander cells.


Subject(s)
Autophagy , HIV Infections/enzymology , HIV-1/physiology , Macrophages/virology , Sodium-Potassium-Exchanging ATPase/biosynthesis , Antiviral Agents/pharmacology , Autophagy/drug effects , Enzyme Induction/drug effects , HIV Infections/pathology , HIV-1/drug effects , Humans , Lipids/chemistry , Models, Biological , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Peptides/pharmacology , RNA Interference , Virus Replication/drug effects
14.
Adv Exp Med Biol ; 1045: 299-320, 2018.
Article in English | MEDLINE | ID: mdl-29896673

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV), also called human herpesvirus-8 (HHV-8), is the eighth human herpesvirus found by Yuan Chang and Patrick Moore, 1992. It is a Rhadinovirus belonging to the gamma herpesvirus subfamily. As known for many gamma herpesviruses, KSHV is also well-correlated to several cancer formations such as Kaposi's sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman's disease. Different from the other herpesvirus subfamily, gamma herpesviruses establish latency as a default infection strategy when they infect to the target cells, as KSHV is present as the latent form in the related cancers. In the latency, the virus expresses a limited number of the genes such as latency-associated nuclear antigen (LANA), v-cyclin (v-CYC, ORF72), v-FLIP (K13), kaposin (K12), and 25 microRNAs (K-miRNAs). The virus replicates according to cellular replication machinery with a viral replication origin (ori-P) and LANA. Then, the replicated genome is segregated equally to daughter cells by appearance to maintain the virus genome copy number per cell. The virus makes the most use of cellular machinery to achieve this end. In this chapter, I would like to review KSHV replication and gene expression in the latency and discuss.


Subject(s)
Genome, Viral , Herpesvirus 8, Human/genetics , Sarcoma, Kaposi/virology , Virus Latency , Virus Replication , Animals , Antigens, Viral/genetics , Antigens, Viral/metabolism , Castleman Disease/virology , Herpesvirus 8, Human/physiology , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
15.
J Med Virol ; 90(4): 753-760, 2018 04.
Article in English | MEDLINE | ID: mdl-29244209

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded viral Fas-associated death domain-like IL-1-converting enzyme inhibitory protein (vFLIP) is one of the latently expressed genes and plays a key role in cell survival and maintenance of latent infection by activating the NF-κB pathway. To obtain a genetic system for studying KSHV vFLIP mutation in the context of the viral genome, we generated recombinant viruses lacking the coding sequence (CDS) of vFLIP gene (K13/ORF71) by bacterial artificial chromosome (BAC) technology and the Escherichia coli Red recombination system. After a series of verification with PCR, restriction digestion and sequencing, the K13 deletion bacmids was transfected into a stable viral producer cell line based on iSLK cells to create vFLIP-knockout mutant. Importantly, human umbilical vein endothelial cells (HUVECs) could be de novo infected by vFLIP mutant virus, which are now available for studying the roles of vFLIP in regulation of other KSHV genes and viral pathogenesis.


Subject(s)
Gene Deletion , Herpesvirus 8, Human/physiology , Viral Proteins/metabolism , Chromosomes, Artificial, Bacterial , Endothelial Cells/virology , Escherichia coli/genetics , Herpesvirus 8, Human/genetics , Human Umbilical Vein Endothelial Cells , Humans , Recombination, Genetic , Viral Proteins/genetics
16.
J Exp Clin Cancer Res ; 36(1): 167, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29179721

ABSTRACT

BACKGROUND: Apigenin is a flavonoid widely distributed in plant kingdom that exerts cytotoxic effects against a variety of solid and haematological cancers. In this study, we investigated the effect of apigenin against primary effusion lymphoma (PEL), a KSHV-associated B cell lymphoma characterized by a very aggressive behavior, displaying constitutive activation of STAT3 as well as of other oncogenic pathways and harboring wtp53. METHODS: Cell death was assessed by trypan blue exclusion assay, FACS analysis as well as by biochemical studies. The latter were also utilized to detect the occurrence of autophagy and the molecular mechanisms leading to the activation of both processes by apigenin. FACS analysis was used to measure the intracellular ROS utilizing DCFDA. RESULTS: We show that apigenin induced PEL cell death and autophagy along with reduction of intracellular ROS. Mechanistically, apigenin activated p53 that induced catalase, a ROS scavenger enzyme, and inhibited STAT3, the most important pro-survival pathway in PEL, as assessed by p53 silencing. On the other hand, STAT3 inhibition by apigenin resulted in p53 activation, since STAT3 negatively influences p53 activity, highlighting a regulatory loop between these two pathways that modulates PEL cell death/survival. CONCLUSION: The findings of this study demonstrate that apigenin may modulate pro-apoptotic and pro-survival pathways representing a valid therapeutic strategy against PEL.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apigenin/pharmacology , Lymphoma, Primary Effusion/metabolism , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis , Autophagy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Primary Effusion/drug therapy , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism
17.
J Virol ; 91(23)2017 12 01.
Article in English | MEDLINE | ID: mdl-28931678

ABSTRACT

Primary effusion lymphoma (PEL) is a lymphogenic disorder associated with Kaposi's sarcoma-associated herpesvirus (KSHV) infection. Key to the survival and proliferation of PEL is the canonical NF-κB pathway, which becomes constitutively activated following overexpression of the viral oncoprotein KSHV vFLIP (ks-vFLIP). This arises from its capacity to form a complex with the modulatory subunit of the IκB kinase (IKK) kinase, IKKγ (or NEMO), resulting in the overproduction of proteins that promote cellular survival and prevent apoptosis, both of which are important drivers of tumorigenesis. Using a combination of cell-based and biophysical assays together with structural techniques, we showed that the observed resistance to cell death is largely independent of autophagy or major death receptor signaling pathways and demonstrated that direct targeting of the ks-vFLIP-IKKγ interaction both in cells and in vitro can be achieved using IKKγ-mimetic peptides. Our results further reveal that these peptides not only induce cell killing but also potently sensitize PEL to the proapoptotic agents tumor necrosis factor alpha and etoposide and are the first to confirm ks-vFLIP as a tractable target for the treatment of PEL and related disorders.IMPORTANCE KSHV vFLIP (ks-vFLIP) has been shown to have a crucial role in cellular transformation, in which it is vital for the survival and proliferation of primary effusion lymphoma (PEL), an aggressive malignancy associated with infection that is resistant to the majority of chemotherapeutic drugs. It operates via subversion of the canonical NF-κB pathway, which requires a physical interaction between ks-vFLIP and the IKK kinase modulatory subunit IKKγ. While this interaction has been directly linked to protection against apoptosis, it is unclear whether the suppression of other cell death pathways implicated in ks-vFLIP pathogenesis is an additional contributor. We demonstrate that the interaction between ks-vFLIP and IKKγ is pivotal in conferring resistance to apoptosis. Additionally, we show that the ks-vFLIP-IKKγ complex can be disrupted using peptides leading to direct killing and the sensitization of PEL cells to proapoptotic agents. Our studies thus provide a framework for future therapeutic interventions.


Subject(s)
Apoptosis , Herpesvirus 8, Human/physiology , I-kappa B Kinase/chemistry , Peptides/metabolism , Peptides/pharmacology , Sarcoma, Kaposi/virology , Autophagy , Etoposide/pharmacology , Herpesvirus 8, Human/chemistry , Humans , I-kappa B Kinase/metabolism , Jurkat Cells , Molecular Mimicry , Peptides/chemistry , Protein Binding , Sarcoma, Kaposi/physiopathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Viral Proteins/metabolism
18.
Virus Genes ; 53(4): 522-531, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28425034

ABSTRACT

The molluscum contagiosum virus (MCV) uses a variety of immune evasion strategies to antagonize host immune responses. Two MCV proteins, MC159 and MC160, contain tandem death effector domains (DEDs). They are reported to inhibit innate immune signaling events such as NF-κB and IRF3 activation, and apoptosis. The RxDL motif of MC159 is required for inhibition of both apoptosis and NF-κB activation. However, the role of the conserved RxDL motif in the MC160 DEDs remained unknown. To answer this question, we performed alanine mutations to neutralize the arginine and aspartate residues present in the MC160 RxDL in both DED1 and DED2. These mutations were further modeled against the structure of the MC159 protein. Surprisingly, the RxDL motif was not required for MC160's ability to inhibit MAVS-induced IFNß activation. Further, unlike previous results with the MC159 protein, mutations within the RxDL motif of MC160 had no effect on the ability of MC160 to dampen TNF-α-induced NF-κB activation. Molecular modeling predictions revealed no overall changes to the structure in the MC160 protein when the amino acids of both RxDL motifs were mutated to alanine (DED1 = R67A D69A; DED2 = R160A D162A). Taken together, our results demonstrate that the RxDL motifs present in the MC160 DEDs are not required for known functions of the viral protein.


Subject(s)
Immune Evasion , Molluscum Contagiosum/virology , Molluscum contagiosum virus/immunology , Viral Proteins/chemistry , Viral Proteins/immunology , Amino Acid Motifs , Apoptosis , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Molluscum Contagiosum/genetics , Molluscum Contagiosum/immunology , Molluscum Contagiosum/physiopathology , Molluscum contagiosum virus/chemistry , Molluscum contagiosum virus/genetics , Protein Domains , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Viral Proteins/genetics
19.
Virology ; 501: 119-126, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27912080

ABSTRACT

Expression of Kaposi's sarcoma herpesvirus vFLIP, a potent activator of NFkB signaling, promotes latency. Inhibition of NFkB signaling promotes lytic reactivation. We previously reported that lytic inducer, RTA, inhibits vFLIP induced NFkB signaling by inducing the degradation of vFLIP via the proteasome. Here we report that the cellular ubiquitin ligase, Itch, is required for RTA induced degradation of vFLIP. Expression of either Itch targeting shRNA or a dominant negative mutant of the ubiquitin ligase both increased the stability of vFLIP in the presence of RTA. Itch potently ubiquitinated vFLIP in vivo and in vitro. We provide evidence for interaction between RTA, vFLIP and Itch and we identified an RTA resistant mutant of vFLIP that is unable to interact with Itch. These observations contribute to our understanding of how RTA counteracts the activities of vFLIP.


Subject(s)
Herpesviridae Infections/enzymology , Immediate-Early Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Herpesvirus 8, Human/enzymology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/genetics , Protein Binding , Proteolysis , Repressor Proteins/genetics , Trans-Activators/genetics , Ubiquitin-Protein Ligases/genetics , Viral Proteins/genetics
20.
Mol Cell ; 64(2): 236-250, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27746017

ABSTRACT

Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , Caspase 8/chemistry , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Fas-Associated Death Domain Protein/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Apoptosis/drug effects , Binding Sites , CARD Signaling Adaptor Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cryoelectron Microscopy , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Death Effector Domain , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression , Humans , Jurkat Cells , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Viral Proteins/genetics , Viral Proteins/metabolism , fas Receptor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL