Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nucleic Acids Res ; 52(4): 1814-1829, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38180827

RESUMO

To establish lifelong, latent infection, herpesviruses circularize their linear, double-stranded, DNA genomes through an unknown mechanism. Kaposi's sarcoma (KS) herpesvirus (KSHV), a gamma herpesvirus, is tightly linked with KS, primary effusion lymphoma, and multicentric Castleman's disease. KSHV persists in latently infected cells as a multi-copy, extrachromosomal episome. Here, we show the KSHV genome rapidly circularizes following infection, and viral protein expression is unnecessary for this process. The DNA damage response (DDR) kinases, ATM and DNA-PKcs, each exert roles, and absence of both severely compromises circularization and latency. These deficiencies were rescued by expression of ATM and DNA-PKcs, but not catalytically inactive mutants. In contrast, γH2AX did not function in KSHV circularization. The linear viral genomic ends resemble a DNA double strand break, and non-homologous DNA end joining (NHEJ) and homologous recombination (HR) reporters indicate both NHEJ and HR contribute to KSHV circularization. Last, we show, similar to KSHV, ATM and DNA-PKcs have roles in circularization of the alpha herpesvirus, herpes simplex virus-1 (HSV-1), while γH2AX does not. Therefore, the DDR mediates KSHV and HSV-1 circularization. This strategy may serve as a general herpesvirus mechanism to initiate latency, and its disruption may provide new opportunities for prevention of herpesvirus disease.


Assuntos
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/genética , Latência Viral/genética , DNA , Reparo do DNA
2.
Life Sci Alliance ; 6(1)2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36375840

RESUMO

Although recent advances in gene therapy provide hope for spinal muscular atrophy (SMA) patients, the pathology remains the leading genetic cause of infant mortality. SMA is a monogenic pathology that originates from the loss of the SMN1 gene in most cases or mutations in rare cases. Interestingly, several SMN1 mutations occur within the TUDOR methylarginine reader domain of SMN. We hypothesized that in SMN1 mutant cases, SMA may emerge from aberrant protein-protein interactions between SMN and key neuronal factors. Using a BioID proteomic approach, we have identified and validated a number of SMN-interacting proteins, including fragile X mental retardation protein (FMRP) family members (FMRFM). Importantly, SMA-linked SMNTUDOR mutant forms (SMNST) failed to interact with FMRFM In agreement with the recent work, we define biochemically that SMN forms droplets in vitro and these droplets are stabilized by RNA, suggesting that SMN could be involved in the formation of membraneless organelles, such as Cajal nuclear bodies. Finally, we found that SMN and FMRP co-fractionate with polysomes, in an RNA-dependent manner, suggesting a potential role in localized translation in motor neurons.


Assuntos
Proteína do X Frágil da Deficiência Intelectual , Atrofia Muscular Espinal , Proteína 1 de Sobrevivência do Neurônio Motor , Humanos , Lactente , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Proteômica , RNA/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética
3.
Nucleic Acids Res ; 49(22): 12895-12911, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34850113

RESUMO

Mixed lineage leukemia 1 (MLL1) is a histone methyltransferase. Kaposi's sarcoma-associated herpesvirus (KSHV) is a leading cause of malignancy in AIDS. KSHV latently infects tumor cells and its genome is decorated with epigenetic marks. Here, we show that KSHV latency-associated nuclear antigen (LANA) recruits MLL1 to viral DNA where it establishes H3K4me3 modifications at the extensive KSHV terminal repeat elements during primary infection. LANA interacts with MLL1 complex members, including WDR5, integrates into the MLL1 complex, and regulates MLL1 activity. We describe the 1.5-Å crystal structure of N-terminal LANA peptide complexed with MLL1 complex member WDR5, which reveals a potential regulatory mechanism. Disruption of MLL1 expression rendered KSHV latency establishment highly deficient. This deficiency was rescued by MLL1 but not by catalytically inactive MLL1. Therefore, MLL1 is LANA regulable and exerts a central role in virus infection. These results suggest broad potential for MLL1 regulation, including by non-host factors.


Assuntos
Antígenos Virais/genética , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Histona-Lisina N-Metiltransferase/genética , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas Nucleares/genética , Sarcoma de Kaposi/genética , Latência Viral/genética , Antígenos Virais/química , Antígenos Virais/metabolismo , Linhagem Celular Tumoral , Cristalografia por Raios X , DNA Viral/genética , DNA Viral/metabolismo , Técnicas de Silenciamento de Genes , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/fisiologia , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/metabolismo , Interações Hospedeiro-Patógeno/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína de Leucina Linfoide-Mieloide/química , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Ligação Proteica , Conformação Proteica , Sarcoma de Kaposi/virologia
4.
Nucleic Acids Res ; 48(21): 11890-11912, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33068409

RESUMO

Eukaryotic cells compartmentalize their internal milieu in order to achieve specific reactions in time and space. This organization in distinct compartments is essential to allow subcellular processing of regulatory signals and generate specific cellular responses. In the nucleus, genetic information is packaged in the form of chromatin, an organized and repeated nucleoprotein structure that is a source of epigenetic information. In addition, cells organize the distribution of macromolecules via various membrane-less nuclear organelles, which have gathered considerable attention in the last few years. The macromolecular multiprotein complexes known as Promyelocytic Leukemia Nuclear Bodies (PML NBs) are an archetype for nuclear membrane-less organelles. Chromatin interactions with nuclear bodies are important to regulate genome function. In this review, we will focus on the dynamic interplay between PML NBs and chromatin. We report how the structure and formation of PML NBs, which may involve phase separation mechanisms, might impact their functions in the regulation of chromatin dynamics. In particular, we will discuss how PML NBs participate in the chromatinization of viral genomes, as well as in the control of specific cellular chromatin assembly pathways which govern physiological mechanisms such as senescence or telomere maintenance.


Assuntos
Núcleo Celular/metabolismo , Cromatina/metabolismo , Genoma Viral , Corpos de Inclusão Intranuclear/metabolismo , Proteína da Leucemia Promielocítica/genética , Processamento de Proteína Pós-Traducional , Núcleo Celular/genética , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Senescência Celular , Cromatina/química , Cromatina/ultraestrutura , Montagem e Desmontagem da Cromatina , Genoma Humano , Histonas/genética , Histonas/metabolismo , Interações Hospedeiro-Patógeno/genética , Humanos , Corpos de Inclusão Intranuclear/química , Corpos de Inclusão Intranuclear/ultraestrutura , Proteína da Leucemia Promielocítica/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Homeostase do Telômero , Vírus/genética , Vírus/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(36): 22443-22451, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32820070

RESUMO

Viruses modulate biochemical cellular pathways to permit infection. A recently described mechanism mediates selective protein interactions between acidic domain readers and unacetylated, lysine-rich regions, opposite of bromodomain function. Kaposi´s sarcoma (KS)-associated herpesvirus (KSHV) is tightly linked with KS, primary effusion lymphoma, and multicentric Castleman's disease. KSHV latently infects cells, and its genome persists as a multicopy, extrachromosomal episome. During latency, KSHV expresses a small subset of genes, including the latency-associated nuclear antigen (LANA), which mediates viral episome persistence. Here we show that LANA contains two tandem, partially overlapping, acidic domain sequences homologous to the SET oncoprotein acidic domain reader. This domain selectively interacts with unacetylated p53, as evidenced by reduced LANA interaction after overexpression of CBP, which acetylates p53, or with an acetylation mimicking carboxyl-terminal domain p53 mutant. Conversely, the interaction of LANA with an acetylation-deficient p53 mutant is enhanced. Significantly, KSHV LANA mutants lacking the acidic domain reader sequence are deficient for establishment of latency and persistent infection. This deficiency was confirmed under physiological conditions, on infection of mice with a murine gammaherpesvirus 68 chimera expressing LANA, where the virus was highly deficient in establishing latent infection in germinal center B cells. Therefore, LANA's acidic domain reader is critical for viral latency. These results implicate an acetylation-dependent mechanism mediating KSHV persistence and expand the role of acidic domain readers.


Assuntos
Antígenos Virais/genética , Antígenos Virais/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Acetilação , Animais , Antígenos Virais/química , DNA Viral/genética , Feminino , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/química , Plasmídeos/genética
6.
Methods Mol Biol ; 2060: 185-197, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617179

RESUMO

Fluorescence in situ hybridization (FISH) has been widely used to analyze genome loci at a single cell level in order to determine within a cell population potential discrepancies in their regulation according to the nuclear positioning. Latent herpes simplex virus 1 (HSV-1) genome remains as an episome in the nucleus of the infected neurons. Accordingly, depending on the location of the viral genomes in the nucleus, they could be targeted by different types of epigenetic regulations important for the establishment and stability of latency, and ultimately for the capacity of HSV-1 to reactivate. Therefore, it is important to take into consideration the interaction of the viral genomes with the nuclear environment to integrate this aspect in the overall set of physiological, immunological, and molecular data that have been produced, and which constitute the main knowledge regarding the biology of HSV-1. In this method chapter we describe in detail the procedure to perform FISH for the detection of HSV-1 genomes particularly during latency and also the combination of this approach with the detection of cellular and/or viral proteins.


Assuntos
Núcleo Celular/virologia , Genoma Viral , Herpesvirus Humano 1/fisiologia , Hibridização in Situ Fluorescente , Neurônios/virologia , Latência Viral , Animais , Núcleo Celular/metabolismo , Humanos , Camundongos , Neurônios/metabolismo
7.
J Virol ; 93(24)2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31554680

RESUMO

Ocular herpes simplex keratitis (HSK) is a consequence of viral reactivations from trigeminal ganglia (TG) and occurs almost exclusively in the same eye in humans. In our murine oro-ocular (OO) model, herpes simplex virus 1 (HSV-1) inoculation in one side of the lip propagates virus to infect the ipsilateral TG. Replication here allows infection of the brainstem and infection of the contralateral TG. Interestingly, HSK was observed in our OO model only from the eye ipsilateral to the site of lip infection. Thus, unilateral restriction of HSV-1 may be due to differential kinetics of virus arrival in the ipsilateral versus contralateral TG. We inoculated mice with HSV-1 reporter viruses and then superinfected them to monitor changes in acute- and latent-phase gene expression in TG after superinfection compared to the control (single inoculation). Delaying superinfection by 4 days after initial right lip inoculation elicited failed superinfecting-virus gene expression and eliminated clinical signs of disease. Initial inoculation with thymidine kinase-deficient HSV-1 (TKdel) completely abolished reactivation of wild-type (WT) superinfecting virus from TG during the latent stage. In light of these seemingly failed infections, viral genome was detected in both TG. Our data demonstrate that inoculation of HSV-1 in the lip propagates virus to both TG, but with delay in reaching the TG contralateral to the side of lip infection. This delay is responsible for restricting viral replication to the ipsilateral TG, which abrogates ocular disease and viral reactivations from the contralateral side. These observations may help to understand why HSK is observed unilaterally in humans, and they provide insight into vaccine strategies to protect against HSK.IMPORTANCE Herpetic keratitis (HK) is the leading cause of blindness by an infectious agent in the developed world. This disease can occur after reactivation of herpes simplex virus 1 in the trigeminal ganglia, leading to dissemination of virus to, and infection of, the cornea. A clinical paradox is evidenced by the bilateral presence of latent viral genomes in both trigeminal ganglia, while for any given patient the disease is unilateral with recurrences in a single eye. Our study links the kinetics of early infection to unilateral disease phenomenon and demonstrates protection against viral reactivation when kinetics are exploited. Our results have direct implications in the understanding of human disease pathogenesis and immunotherapeutic strategies for the treatment of HK and viral reactivations.


Assuntos
Herpesvirus Humano 1/fisiologia , Ceratite Herpética/virologia , Lábio/virologia , Latência Viral/fisiologia , Replicação Viral/fisiologia , Animais , Córnea/virologia , Feminino , Regulação Viral da Expressão Gênica , Genes Virais/genética , Herpesvirus Humano 1/genética , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/metabolismo , Gânglio Trigeminal/virologia , Latência Viral/genética
8.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626680

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA) is a 1,162-amino-acid protein that mediates episome persistence of viral genomes. LANA binds the KSHV terminal-repeat (TR) sequence through its carboxy-terminal domain to mediate DNA replication. LANA simultaneously binds mitotic chromosomes and TR DNA to segregate virus genomes to daughter cell nuclei. Amino-terminal LANA attaches to chromosomes by binding histones H2A/H2B, and carboxy-terminal LANA contributes to mitotic-chromosome binding. Although amino- and carboxy-terminal LANA are essential for episome persistence, they are not sufficient, since deletion of all internal LANA sequence renders LANA highly deficient for episome maintenance. Internal LANA sequence upstream of the internal repeat elements contributes to episome segregation and persistence. Here, we investigate this region with a panel of LANA deletion mutants. Mutants retained the ability to associate with mitotic chromosomes and bind TR DNA. In contrast to prior results, deletion of most of this sequence did not reduce LANA's ability to mediate DNA replication. Deletions of upstream sequence within the region compromised segregation of TR DNA to daughter cells, as assessed by retention of green fluorescent protein (GFP) expression from a replication-deficient TR plasmid. However, deletion of this upstream sequence did not reduce episome maintenance. In contrast, deletions that included an 80-amino-acid sequence immediately downstream resulted in highly deficient episome persistence. LANA with this downstream sequence deleted maintained the ability to replicate and segregate TR DNA, suggesting a unique role for the residues. Therefore, this work identifies adjacent LANA regions with distinct roles in episome segregation and persistence.IMPORTANCE KSHV LANA mediates episomal persistence of viral genomes. LANA binds the KSHV terminal-repeat (TR) sequence to mediate DNA replication and tethers KSHV DNA to mitotic chromosomes to segregate genomes to daughter cell nuclei. Here, we investigate LANA sequence upstream of the internal repeat elements that contributes to episome segregation and persistence. Mutants with deletions within this sequence maintained the ability to bind mitotic chromosomes or bind and replicate TR DNA. Deletion of upstream sequence within the region reduced segregation of TR DNA to daughter cells, but not episome maintenance. In contrast, mutants with deletions of 80 amino acids immediately downstream were highly deficient for episome persistence yet maintained the ability to replicate and segregate TR DNA, the two principal components of episome persistence, suggesting another role for the residues. In summary, this work identifies adjacent LANA sequence with distinct roles in episome segregation and persistence.


Assuntos
Antígenos Virais/genética , Herpesvirus Humano 8/genética , Proteínas Nucleares/genética , Plasmídeos/genética , Sarcoma de Kaposi/virologia , Antígenos Nucleares/genética , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/virologia , Cromossomos/genética , Replicação do DNA/genética , DNA Viral/genética , Genoma Viral/genética , Células HEK293 , Humanos , Mitose/genética , Sequências Repetidas Terminais/genética , Proteínas Virais/genética , Latência Viral/genética , Replicação Viral/genética
9.
Sci Rep ; 6: 39144, 2016 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-28004818

RESUMO

Cancer cells have been increasingly grown in pharmaceutical research to understand tumorigenesis and develop new therapeutic drugs. Currently, cells are typically grown using two-dimensional (2-D) cell culture approaches, where the native tumor microenvironment is difficult to recapitulate. Thus, one of the main obstacles in oncology is the lack of proper infection models that recount main features present in tumors. In recent years, microtechnology-based platforms have been employed to generate three-dimensional (3-D) models that better mimic the native microenvironment in cell culture. Here, we present an innovative approach to culture Kaposi's sarcoma-associated herpesvirus (KSHV) infected human B cells in 3-D using a microwell array system. The results demonstrate that the KSHV-infected B cells can be grown up to 15 days in a 3-D culture. Compared with 2-D, cells grown in 3-D had increased numbers of KSHV latency-associated nuclear antigen (LANA) dots, as detected by immunofluorescence microscopy, indicating a higher viral genome copy number. Cells in 3-D also demonstrated a higher rate of lytic reactivation. The 3-D microwell array system has the potential to improve 3-D cell oncology models and allow for better-controlled studies for drug discovery.


Assuntos
Antígenos Virais/genética , Linfócitos B/virologia , Técnicas de Cultura de Células/instrumentação , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/fisiologia , Proteínas Nucleares/genética , Antígenos Virais/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas Nucleares/metabolismo , Carga Viral , Ativação Viral , Latência Viral/genética
10.
Front Microbiol ; 7: 1149, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27570517

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) has an etiologic role in Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. These diseases are most common in immunocompromised individuals, especially those with AIDS. Similar to all herpesviruses, KSHV infection is lifelong. KSHV infection in tumor cells is primarily latent, with only a small subset of cells undergoing lytic infection. During latency, the KSHV genome persists as a multiple copy, extrachromosomal episome in the nucleus. In order to persist in proliferating tumor cells, the viral genome replicates once per cell cycle and then segregates to daughter cell nuclei. KSHV only expresses several genes during latent infection. Prominent among these genes, is the latency-associated nuclear antigen (LANA). LANA is responsible for KSHV genome persistence and also exerts transcriptional regulatory effects. LANA mediates KSHV DNA replication and in addition, is responsible for segregation of replicated genomes to daughter nuclei. LANA serves as a molecular tether, bridging the viral genome to mitotic chromosomes to ensure that KSHV DNA reaches progeny nuclei. N-terminal LANA attaches to mitotic chromosomes by binding histones H2A/H2B at the surface of the nucleosome. C-terminal LANA binds specific KSHV DNA sequence and also has a role in chromosome attachment. In addition to the essential roles of N- and C-terminal LANA in genome persistence, internal LANA sequence is also critical for efficient episome maintenance. LANA's role as an essential mediator of virus persistence makes it an attractive target for inhibition in order to prevent or treat KSHV infection and disease.

11.
J Virol ; 90(17): 7667-83, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27307564

RESUMO

UNLABELLED: Viruses have evolved mechanisms to hijack components of cellular E3 ubiquitin ligases, thus modulating the ubiquitination pathway. However, the biological relevance of such mechanisms for viral pathogenesis in vivo remains largely unknown. Here, we utilized murid herpesvirus 4 (MuHV-4) infection of mice as a model system to address the role of MuHV-4 latency-associated nuclear antigen (mLANA) E3 ligase activity in gammaherpesvirus latent infection. We show that specific mutations in the mLANA SOCS box (V199A, V199A/L202A, or P203A/P206A) disrupted mLANA's ability to recruit Elongin C and Cullin 5, thereby impairing the formation of the Elongin BC/Cullin 5/SOCS (EC5S(mLANA)) complex and mLANA's E3 ligase activity on host NF-κB and Myc. Although these mutations resulted in considerably reduced mLANA binding to viral terminal repeat DNA as assessed by electrophoretic mobility shift assay (EMSA), the mutations did not disrupt mLANA's ability to mediate episome persistence. In vivo, MuHV-4 recombinant viruses bearing these mLANA SOCS box mutations exhibited a deficit in latency amplification in germinal center (GC) B cells. These findings demonstrate that the E3 ligase activity of mLANA contributes to gammaherpesvirus-driven GC B cell proliferation. Hence, pharmacological inhibition of viral E3 ligase activity through targeting SOCS box motifs is a putative strategy to control gammaherpesvirus-driven lymphoproliferation and associated disease. IMPORTANCE: The gammaherpesviruses Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) cause lifelong persistent infection and play causative roles in several human malignancies. Colonization of B cells is crucial for virus persistence, and access to the B cell compartment is gained by virus-driven proliferation in germinal center (GC) B cells. Infection of B cells is predominantly latent, with the viral genome persisting as a multicopy episome and expressing only a small subset of viral genes. Here, we focused on latency-associated nuclear antigen (mLANA) encoded by murid herpesvirus-4 (MuHV-4), which exhibits homology in sequence, structure, and function to KSHV LANA (kLANA), thereby allowing the study of LANA-mediated pathogenesis in mice. Our experiments show that mLANA's E3 ubiquitin ligase activity is necessary for efficient expansion of latency in GC B cells, suggesting that the development of pharmacological inhibitors of LANA E3 ubiquitin ligase activity may allow strategies to interfere with gammaherpesvirus-driven lymphoproliferation and associated disease.


Assuntos
Antígenos Virais/metabolismo , Linfócitos B/fisiologia , Proliferação de Células , Centro Germinativo/citologia , Interações Hospedeiro-Patógeno , Proteínas Nucleares/metabolismo , Rhadinovirus/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antígenos Virais/genética , Análise Mutacional de DNA , DNA Viral/metabolismo , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Proteínas Nucleares/genética , Ligação Proteica
12.
Biosens Bioelectron ; 80: 230-236, 2016 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26851580

RESUMO

Certain viruses, such as herpesviruses, are capable of persistent and latent infection of host cells. Distinguishing and separating live, latently infected cells from uninfected cells is not easily attainable using current approaches. The ability to perform such separation would greatly enhance the ability to study primary, infected cells and potentially enable elimination of latently infected cells from the host. Here, the dielectrophoretic response of B cells infected with Kaposi's sarcoma-associated herpesvirus (KSHV) were investigated and compared to uninfected B cells. We evaluated the effect of applied voltage, signal frequency, and flow rate of the sample on the cell capture efficiency. We achieved 37.1% ± 8.5% difference in capture efficiencies between latently KSHV-infected and uninfected BJAB B lymphoma cells at the chip operational conditions of 1V, 50 kHz and 0.02 µl/min sample flow rate. Our results show that latently infected B lymphoma cells demonstrated significantly different electrical response compared to uninfected B cells and DEP-based microchips can be potentially used for sorting latently infected cells based on their electrical properties.


Assuntos
Técnicas Biossensoriais , Herpesvirus Humano 8/isolamento & purificação , Linfoma de Células B/virologia , Proteínas Virais/isolamento & purificação , Herpesvirus Humano 8/patogenicidade , Humanos , Linfoma de Células B/diagnóstico , Sarcoma de Kaposi/virologia , Proteínas Virais/química
13.
J Biol Chem ; 290(47): 28084-28096, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26420481

RESUMO

Kaposi sarcoma-associated herpesvirus (KSHV) has a causative role in several human malignancies. KSHV latency-associated nuclear antigen (LANA) mediates persistence of viral episomes in latently infected cells. LANA mediates KSHV DNA replication and segregates episomes to progeny nuclei. The structure of the LANA DNA binding domain was recently solved, revealing a positive electrostatic patch opposite the DNA binding surface, which is the site of BET protein binding. Here we investigate the functional role of the positive patch in LANA-mediated episome persistence. As expected, LANA mutants with alanine or glutamate substitutions in the central, peripheral, or lateral portions of the positive patch maintained the ability to bind DNA by EMSA. However, all of the substitution mutants were deficient for LANA DNA replication and episome maintenance. Mutation of the peripheral region generated the largest deficiencies. Despite these deficiencies, all positive patch mutants concentrated to dots along mitotic chromosomes in cells containing episomes, similar to LANA. The central and peripheral mutants, but not the lateral mutants, were reduced for BET protein interaction as assessed by co-immunoprecipitation. However, defects in BET protein binding were independent of episome maintenance function. Overall, the reductions in episome maintenance closely correlated with DNA replication deficiencies, suggesting that the replication defects account for the reduced episome persistence. Therefore, the electrostatic patch exerts a key role in LANA-mediated DNA replication and episome persistence and may act through a host cell partner(s) other than a BET protein or by inducing specific structures or complexes.


Assuntos
Antígenos Virais/metabolismo , Replicação do DNA , Herpesvirus Humano 8/genética , Proteínas Nucleares/metabolismo , Plasmídeos/fisiologia , Latência Viral , Sítios de Ligação , Linhagem Celular Tumoral , Herpesvirus Humano 8/imunologia , Humanos , Eletricidade Estática , Sequências Repetidas Terminais
14.
Nucleic Acids Res ; 43(20): 10039-54, 2015 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-26424851

RESUMO

Latency-associated nuclear antigen (LANA) is central to episomal tethering, replication and transcriptional regulation of γ2-herpesviruses. LANA binds cooperatively to the terminal repeat (TR) region of the viral episome via adjacent LANA binding sites (LBS), but the molecular mechanism by which LANA assembles on the TR remains elusive. We show that KSHV LANA and MHV-68 LANA proteins bind LBS DNA using strikingly different modes. Solution structure of LANA complexes revealed that while kLANA tetramer is intrinsically bent both in the free and bound state to LBS1-2 DNA, mLANA oligomers instead adopt a rigid linear conformation. In addition, we report a novel non-ring kLANA structure that displays more flexibility at its assembly interface than previously demonstrated. We identified a hydrophobic pivot point located at the dimer-dimer assembly interface, which gives rotational freedom for kLANA to adopt variable conformations to accommodate both LBS1-2 and LBS2-1-3 DNA. Alterations in the arrangement of LBS within TR or at the tetramer assembly interface have a drastic effect on the ability of kLANA binding. We also show kLANA and mLANA DNA binding functions can be reciprocated. Although KSHV and MHV-68 are closely related, the findings provide new insights into how the structure, oligomerization, and DNA binding of LANA have evolved differently to assemble on the TR DNA.


Assuntos
Antígenos Virais/química , DNA Viral/química , Herpesvirus Humano 8 , Proteínas Nucleares/química , Rhadinovirus , Antígenos Virais/genética , Antígenos Virais/metabolismo , Sítios de Ligação , DNA Viral/metabolismo , Modelos Moleculares , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Conformação de Ácido Nucleico , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Sequências Repetidas Terminais , Termodinâmica
15.
Proc Natl Acad Sci U S A ; 112(32): E4354-63, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26195743

RESUMO

Recent advances in biosensing technologies present great potential for medical diagnostics, thus improving clinical decisions. However, creating a label-free general sensing platform capable of detecting multiple biotargets in various clinical specimens over a wide dynamic range, without lengthy sample-processing steps, remains a considerable challenge. In practice, these barriers prevent broad applications in clinics and at patients' homes. Here, we demonstrate the nanoplasmonic electrical field-enhanced resonating device (NE(2)RD), which addresses all these impediments on a single platform. The NE(2)RD employs an immunodetection assay to capture biotargets, and precisely measures spectral color changes by their wavelength and extinction intensity shifts in nanoparticles without prior sample labeling or preprocessing. We present through multiple examples, a label-free, quantitative, portable, multitarget platform by rapidly detecting various protein biomarkers, drugs, protein allergens, bacteria, eukaryotic cells, and distinct viruses. The linear dynamic range of NE(2)RD is five orders of magnitude broader than ELISA, with a sensitivity down to 400 fg/mL This range and sensitivity are achieved by self-assembling gold nanoparticles to generate hot spots on a 3D-oriented substrate for ultrasensitive measurements. We demonstrate that this precise platform handles multiple clinical samples such as whole blood, serum, and saliva without sample preprocessing under diverse conditions of temperature, pH, and ionic strength. The NE(2)RD's broad dynamic range, detection limit, and portability integrated with a disposable fluidic chip have broad applications, potentially enabling the transition toward precision medicine at the point-of-care or primary care settings and at patients' homes.


Assuntos
Técnicas Biossensoriais/instrumentação , Técnicas e Procedimentos Diagnósticos/instrumentação , Eletricidade , Nanoestruturas/química , Linhagem Celular Tumoral , Coinfecção/diagnóstico , Meio Ambiente , Ensaio de Imunoadsorção Enzimática , Desenho de Equipamento , Humanos , Concentração de Íons de Hidrogênio , Limite de Detecção , Microfluídica , Concentração Osmolar , Reprodutibilidade dos Testes , Temperatura
16.
Sci Rep ; 5: 9919, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26046668

RESUMO

We report a biosensing platform for viral load measurement through electrical sensing of viruses on a flexible plastic microchip with printed electrodes. Point-of-care (POC) viral load measurement is of paramount importance with significant impact on a broad range of applications, including infectious disease diagnostics and treatment monitoring specifically in resource-constrained settings. Here, we present a broadly applicable and inexpensive biosensing technology for accurate quantification of bioagents, including viruses in biological samples, such as plasma and artificial saliva, at clinically relevant concentrations. Our microchip fabrication is simple and mass-producible as we print microelectrodes on flexible plastic substrates using conductive inks. We evaluated the microchip technology by detecting and quantifying multiple Human Immunodeficiency Virus (HIV) subtypes (A, B, C, D, E, G, and panel), Epstein-Barr Virus (EBV), and Kaposi's Sarcoma-associated Herpes Virus (KSHV) in a fingerprick volume (50 µL) of PBS, plasma, and artificial saliva samples for a broad range of virus concentrations between 10(2) copies/mL and 10(7) copies/mL. We have also evaluated the microchip platform with discarded, de-identified HIV-infected patient samples by comparing our microchip viral load measurement results with reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) as the gold standard method using Bland-Altman Analysis.


Assuntos
Infecções por HIV/diagnóstico , HIV-1/isolamento & purificação , Herpesvirus Humano 4/isolamento & purificação , Herpesvirus Humano 8/isolamento & purificação , Dispositivos Lab-On-A-Chip/normas , Carga Viral , Infecções por Vírus Epstein-Barr/sangue , Infecções por Vírus Epstein-Barr/diagnóstico , Infecções por Vírus Epstein-Barr/virologia , Infecções por HIV/sangue , Infecções por HIV/virologia , HIV-1/fisiologia , Infecções por Herpesviridae/sangue , Infecções por Herpesviridae/diagnóstico , Infecções por Herpesviridae/virologia , Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 8/fisiologia , Humanos , Sistemas Automatizados de Assistência Junto ao Leito , Padrões de Referência
17.
Proc Natl Acad Sci U S A ; 111(32): 11816-21, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25071216

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) latently infects tumor cells and persists as a multiple-copy, extrachromosomal, circular episome. To persist, the viral genome must replicate with each cell cycle. The KSHV latency-associated nuclear antigen (LANA) mediates viral DNA replication and persistence, but little is known regarding the underlying mechanisms. We find that LANA recruits replication factor C (RFC), the DNA polymerase clamp [proliferating cell nuclear antigen (PCNA)] loader, to drive DNA replication efficiently. Mutated LANA lacking RFC interaction was deficient for LANA-mediated DNA replication and episome persistence. RFC depletion had a negative impact on LANA's ability to replicate and maintain viral DNA in cells containing artificial KSHV episomes or in infected cells, leading to loss of virus. LANA substantially increased PCNA loading onto DNA in vitro and recruited RFC and PCNA to KSHV DNA in cells. These findings suggest that PCNA loading is a rate-limiting step in DNA replication that is incompatible with viral survival. LANA enhancement of PCNA loading permits efficient virus replication and persistence, revealing a previously unidentified mechanism for KSHV latency.


Assuntos
Antígenos Virais/fisiologia , Herpesvirus Humano 8/fisiologia , Herpesvirus Humano 8/patogenicidade , Proteínas Nucleares/fisiologia , Proteína de Replicação C/fisiologia , Replicação Viral/fisiologia , Linhagem Celular Tumoral , Replicação do DNA/fisiologia , DNA Viral/biossíntese , DNA Viral/genética , DNA Polimerase Dirigida por DNA/fisiologia , Técnicas de Silenciamento de Genes , Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno , Humanos , Antígeno Nuclear de Célula em Proliferação/fisiologia , Proteína de Replicação C/antagonistas & inibidores , Proteína de Replicação C/genética , Sarcoma de Kaposi/fisiopatologia , Sarcoma de Kaposi/virologia , Latência Viral/fisiologia
18.
Virology ; 448: 344-55, 2014 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-24314665

RESUMO

Kaposi's sarcoma-associated herpesvirus LANA (1162 residues) mediates episomal persistence of viral genomes during latency. LANA mediates viral DNA replication and segregates episomes to daughter nuclei. A 59 residue deletion immediately upstream of the internal repeat elements rendered LANA highly deficient for DNA replication and modestly deficient for the ability to segregate episomes, while smaller deletions did not. The 59 amino acid deletion reduced LANA episome persistence by ~14-fold, while sequentially smaller deletions resulted in ~3-fold, or no deficiency. Three distinct LANA regions reorganized heterochromatin, one of which contains the deleted sequence, but the deletion did not abolish LANA's ability to alter chromatin. Therefore, this work identifies a short internal LANA sequence that is critical for DNA replication, has modest effects on episome segregation, and substantially impacts episome persistence; this region may exert its effects through an interacting host cell protein(s).


Assuntos
Antígenos Virais/química , Antígenos Virais/metabolismo , Replicação do DNA , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Plasmídeos/genética , Sequências Repetidas Terminais , Sequência de Aminoácidos , Antígenos Virais/genética , Linhagem Celular , Cromatina/metabolismo , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/química , Herpesvirus Humano 8/fisiologia , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Replicação Viral
19.
Virologie (Montrouge) ; 17(2): 96-110, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-31910538

RESUMO

During viral infection, the amount of viral mRNAs expressed is not only a reflection of the viral gene transcription level: mRNA stability and nucleo-cytoplasmic export are also important for optimal viral gene expression during the productive cycle. It is now well established that herpesviruses express a protein absolutely required for the cytoplasmic accumulation of some viral mRNAs transcribed from intronless genes. This family of proteins comprises the EB2 factor from Epstein-Barr virus (EBV), and its similar proteins: ICP27 from HSV-1, UL69 from cytomegalovirus (CMV), ORF57 from KSHV and IE4 from VZV. These proteins are able to stabilize their target mRNAs in the nucleus and, by interacting with various cellular factors (TAP/NXF1, SR proteins, RBM15 etc), promote mRNA export to the cytoplasm, where they are also involved in the translation efficiency of these viral mRNAs. On the basis of their essential role in the viral productive cycle, these multifunctional viral factors should be considered as important targets for therapeutic approaches.

20.
Nucleic Acids Res ; 40(14): 6834-49, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22505578

RESUMO

The Epstein-Barr Virus (EBV) protein EB2 (also called Mta, SM and BMLF1), is an essential nuclear protein produced during the replicative cycle of EBV. EB2 is required for the efficient cytoplasmic accumulation of viral mRNAs derived from intronless genes. EB2 is an RNA-binding protein whose expression has been shown to influence RNA stability, splicing, nuclear export and translation. Using a yeast two-hybrid screen, we have identified three SR proteins, SF2/ASF, 9G8 and SRp20, as cellular partners of EB2. Then, by using siRNA to deplete cells of specific SR proteins, we found that SRp20 plays an essential role in the processing of several model mRNAs: the Renilla luciferase reporter mRNA, the human ß-globin cDNA transcript and two EBV late mRNAs. These four mRNAs were previously found to be highly dependent on EB2 for their efficient cytoplasmic accumulation. Here, we show that SRp20 depletion results in an increase in the accumulation of these mRNAs, which correlates with an absence of additive effect of EB2, suggesting that EB2 functions by antagonizing SRp20. Moreover, by using RNA-immunoprecipitation assays we found that EB2 enhances the association of SRp20 with the ß-globin transcript suggesting that EB2 acts by stabilizing SRp20's labile interactions with the RNA.


Assuntos
Fosfoproteínas/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transativadores/metabolismo , Núcleo Celular/virologia , Citoplasma/metabolismo , Citoplasma/virologia , Regulação para Baixo , Células HEK293 , Células HeLa , Humanos , Luciferases de Renilla/genética , Mutação , Proteínas Nucleares/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Domínios e Motivos de Interação entre Proteínas , Proteínas de Ligação a RNA/antagonistas & inibidores , Fatores de Processamento de Serina-Arginina , Transativadores/química , Transativadores/genética , Técnicas do Sistema de Duplo-Híbrido , Globinas beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA